1
|
Sun L, Wang B, Yang D, Zhou W, Tang Y, Li X, Lv H, Hou M. Relationship between platelet distribution width and non-dipping pattern in children with essential hypertension. BMC Pediatr 2025; 25:54. [PMID: 39844072 PMCID: PMC11752664 DOI: 10.1186/s12887-025-05420-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Hypertension has shown a trend of prevalence at younger ages, and the non-dipping pattern is associated with target organ damage in hypertension. However, few studies have yet investigated the clinical characteristics and risk factors of non-dipper status in essential hypertension children. This study aimed to explore the clinical characteristics and possible indicators associated with non-dipper status in children with essential hypertension. METHODS A total of 125 children (99 boys, 26 girls) with untreated essential hypertension were retrospectively included in this study. Non-dipping was defined as a nocturnal drop in systolic or diastolic BP (SBP, DBP) < 10%. Clinical data, ambulatory blood pressure monitoring (ABPM), laboratory and echocardiography parameters were recorded from the hospital database. RESULTS Non-dipping pattern was found in 74 (59.2%) children and the dipping pattern in 51(40.8%) children, and the nocturnal SBP drop was 8.43 ± 0.71 (%), and the DBP drop was 14.44 ± 0.86 (%). The proportion of children with left ventricular hypertrophy was higher in the non-dipping group than in the dipping group. The platelet distribution width, high-sensitivity C-reactive protein (hs-CRP) and triglycerides (TG) levels were higher in the non-dipping group compared with the dipping group. In multivariate logistic regression analysis, PDW, TG and hs-CRP were found to be associated with the non-dipping pattern. CONCLUSION Non-dipping pattern in children hypertension is common, and the proportion of left ventricular hypertrophy is higher in non-dipping hypertension children. Moreover, higher PDW, hs-CRP and TG levels are the risk factors for non-dipping status in essential hypertension children.
Collapse
Affiliation(s)
- Ling Sun
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bo Wang
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Daoping Yang
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wanping Zhou
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yunjia Tang
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuan Li
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Lv
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Hou
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Xu B, Ye X, Wen Z, Chen J, Chen M, Shen M, Xu Y, Wang J, Chen S. Biphasic Effect of Thyroid Hormone on Megakaryopoiesis and Platelet Production. Thyroid 2024. [PMID: 39692608 DOI: 10.1089/thy.2024.0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Background: Abnormal platelet counts are frequently observed in patients with thyroid dysfunction; however, the direct impact of thyroid hormones on thrombopoiesis remains largely undefined. Methods: This study elucidates the dose-response effect of the thyroid hormone triiodothyronine (T3) on megakaryocyte (MK) development and thrombopoiesis using both a murine model of hyperthyroidism/hypothyroidism and in vitro cultures of human cord blood CD34+ cell-derived MKs. After the application of inhibitors to MKs, the examination of total and phosphorylated protein levels of the phosphoinositide 3-kinase (PI3K)/AKT pathway was utilized to assess the specific mechanisms of T3 action. The use of autophagy dual-staining lentivirus and transmission electron microscopy was employed to evaluate the impact of T3 on the autophagy flux in MKs. Mouse whole-body irradiation and bone marrow transplantation models are applied to assess the influence of T3 on the recovery of MKs/platelets in vivo. Results: We found that physiological or slightly elevated thyroid hormone levels are essential for sustaining MK development and thrombopoiesis, primarily through the TRα-PI3K/AKT signaling pathway. In contrast, supraphysiological thyroid hormone concentrations induce MK apoptosis via excessive autophagy, thereby reducing platelet production. Conclusions: Here, we present evidence that the thyroid hormone influences MK development and platelet production in a concentration-dependent manner, exhibiting a dualistic role. Our discoveries shed new light on the intricate relationship between thyroid hormones and platelet formation, offering novel perspectives on the pathophysiological consequences of thyroid disorders.
Collapse
Affiliation(s)
- Baichuan Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianpeng Ye
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhaoyang Wen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jun Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mo Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mingqiang Shen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yang Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shilei Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
3
|
Meng P, Liu W, Lao J, Liu X, Zhang Y, Sun Y, Zhou R, Du C, Wang J, Zhao D, Lin Q, Zhang Y. Paclitaxel improves thrombopoiesis in the absence of thrombopoietin receptor (Mpl). J Thromb Haemost 2024; 22:3599-3613. [PMID: 39307245 DOI: 10.1016/j.jtha.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 10/17/2024]
Abstract
BACKGROUND Platelets are critical for thrombosis and hemostasis. The THPO-MPL pathway is the primary pathway for generating thrombocytes. Dysregulation of thrombopoiesis results in platelet formation and/or function-related disorders, such as thrombocytopenia. Paclitaxel is an extensively utilized chemotherapeutic agent and its activity may be related to platelets, but the effect of paclitaxel on thrombocytopoiesis warrants comprehensive exploration. OBJECTIVES We focused on identifying factors that regulate thrombocyte production and elucidating paclitaxel's regulatory mechanisms on thrombocytopoiesis, with a particular emphasis on discovering mechanisms that bypass THPO-MPL pathways. METHODS We performed drug screenings using the Tg(mpl:eGFP) zebrafish model in vivo to identify Food and Drug Administration-approved compounds capable of boosting thrombocyte production. An injury experiment was used to evaluate thrombocyte function. Bromodeoxyuridine assays, terminal deoxynucleotidyl transferase dUTP nick-end labeling, and RNA sequencing analyses were performed to explore cytological and molecular mechanisms. Routine blood testing and flow cytometry were used to analyze mouse phenotypes. RESULTS We found that paclitaxel expands thrombocytes by accelerating the proliferation of thrombocytic lineage cells in zebrafish and elevates platelet levels in mice. This effect occurs by bypassing the thrombopoietin receptor (Mpl). We found that paclitaxel promotes thrombopoiesis, potentially involving the JAK2-ERK1/2 MAPK signaling cascade, a pathway integral to MPL and other regulators. Our results further demonstrate that ERK1/2 is at least partially downstream of JAK2 in paclitaxel-induced thrombopoiesis. CONCLUSION Paclitaxel could promote thrombopoiesis by bypassing Mpl but presumably via the JAK2-ERK1/2 MAPK pathways. It will aid in understanding the relationship between paclitaxel and platelets clinically, and paclitaxel may have potential value for safeguarding platelets and improving thrombocytosis in related diseases.
Collapse
Affiliation(s)
- Panpan Meng
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China; Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenyu Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiawen Lao
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xunwei Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yangping Zhang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Sun
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Riyang Zhou
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Guangdong Higher Education Institutes, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Dejian Zhao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Qing Lin
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China; Department of Hematology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Yiyue Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
4
|
Du XJ, Huang YQ, Li XY, Liao Y, Jin HF, Du JB. Age and mean platelet volume-based nomogram for predicting the therapeutic efficacy of metoprolol in Chinese pediatric patients with vasovagal syncope. World J Pediatr 2024; 20:957-965. [PMID: 38613734 PMCID: PMC11422430 DOI: 10.1007/s12519-024-00802-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/28/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Vasovagal syncope (VVS) is the most common type of orthostatic intolerance in children. We investigated whether platelet-related factors related to treatment efficacy in children suffering from VVS treated with metoprolol. METHODS Metoprolol-treated VVS patients were recruited. The median duration of therapy was three months. Patients were followed and divided into two groups, treament-effective group and treatment-ineffective group. Logistic and least absolute shrinkage selection operator regressions were used to examine treatment outcome variables. Receiver-operating characteristic (ROC) curves, precision-recall (PR) curves, calibration plots, and decision curve analyses were used to evaluate the nomogram model. RESULTS Among the 72 patients who complete the follow-up, treatment-effective group and treatment-ineffective group included 42 (58.3%) and 30 (41.7%) cases, respectively. The patients in the treatment-effective group exhibited higher mean platelet volume (MPV) [(11.0 ± 1.0) fl vs. (9.8 ± 1.0) fl, P < 0.01] and platelet distribution width [12.7% (12.3%, 14.3%) vs. 11.3% (10.2%, 12.2%), P < 0.01] than those in the treatment-ineffective group. The sex ratio was significantly different (P = 0.046). A fit model comprising age [odds ratio (OR) = 0.766, 95% confidence interval (CI) = 0.594-0.987] and MPV (OR = 5.613, 95% CI = 2.297-13.711) might predict therapeutic efficacy. The area under the curve of the ROC and PR curves was computed to be 0.85 and 0.9, respectively. The P value of the Hosmer-Lemeshow test was 0.27. The decision curve analysis confirmed that managing children with VVS based on the predictive model led to a net advantage ranging from 0.01 to 0.58. The nomogram is convenient for clinical applications. CONCLUSION A novel nomogram based on age and MPV can predict the therapeutic benefits of metoprolol in children with VVS.
Collapse
Affiliation(s)
- Xiao-Juan Du
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Ya-Qian Huang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xue-Ying Li
- Department of Statistics, Peking University First Hospital, Beijing, 100034, China
| | - Ying Liao
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China.
| | - Hong-Fang Jin
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing, 100191, China.
| | - Jun-Bao Du
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, No. 38, Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
5
|
Meng X, Liu Z, Deng L, Yang Y, Zhu Y, Sun X, Hao Y, He Y, Fu J. Hydrogen Therapy Reverses Cancer-Associated Fibroblasts Phenotypes and Remodels Stromal Microenvironment to Stimulate Systematic Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401269. [PMID: 38757665 PMCID: PMC11267370 DOI: 10.1002/advs.202401269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/02/2024] [Indexed: 05/18/2024]
Abstract
Tumor microenvironment (TME) plays an important role in the tumor progression. Among TME components, cancer-associated fibroblasts (CAFs) show multiple tumor-promoting effects and can induce tumor immune evasion and drug-resistance. Regulating CAFs can be a potential strategy to augment systemic anti-tumor immunity. Here, the study observes that hydrogen treatment can alleviate intracellular reactive oxygen species of CAFs and reshape CAFs' tumor-promoting and immune-suppressive phenotypes. Accordingly, a controllable and TME-responsive hydrogen therapy based on a CaCO3 nanoparticles-coated magnesium system (Mg-CaCO3) is developed. The hydrogen therapy by Mg-CaCO3 can not only directly kill tumor cells, but also inhibit pro-tumor and immune suppressive factors in CAFs, and thus augment immune activities of CD4+ T cells. As implanted in situ, Mg-CaCO3 can significantly suppress tumor growth, turn the "cold" primary tumor into "hot", and stimulate systematic anti-tumor immunity, which is confirmed by the bilateral tumor transplantation models of "cold tumor" (4T1 cells) and "hot tumor" (MC38 cells). This hydrogen therapy system reverses immune suppressive phenotypes of CAFs, thus providing a systematic anti-tumor immune stimulating strategy by remodeling tumor stromal microenvironment.
Collapse
Affiliation(s)
- Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Liang Deng
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yangzi Yang
- Department of Orthopedic SurgerySpine CenterChangzheng HospitalNavy Medical UniversityNo. 415 Fengyang RoadShanghai200003P. R. China
| | - Yingchun Zhu
- Key Laboratory of Inorganic Coating MaterialsShanghai Institute of CeramicsChinese Academy of SciencesShanghai200050P. R. China
| | - Xiaoying Sun
- College of SciencesShanghai UniversityShanghai200444P. R. China
| | - Yongqiang Hao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- College of StomatologyNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Jiao Tong UniversityShanghai200011P. R. China
| | - Jingke Fu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Clinical and Translational Research Center for 3D Printing TechnologyShanghai Engineering Research Center of Innovative Orthopaedic Instruments and Personalized MedicineShanghai200011P. R. China
| |
Collapse
|
6
|
von Känel R. Stress-Induced Hypercoagulability: Insights from Epidemiological and Mechanistic Studies, and Clinical Integration. Semin Thromb Hemost 2024. [PMID: 38914118 DOI: 10.1055/s-0044-1787660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
By integrating findings from comprehensive reviews, meta-analyses, and cutting-edge genetic studies, this article illuminates the significance of stress-induced hypercoagulability in clinical medicine. In particular, the findings from numerous prospective cohort studies indicate that stress and hemostatic factors of a hypercoagulable state are associated with increased incident risk and poor prognosis for atherosclerotic cardiovascular disease and venous thromboembolism. Mendelian randomization studies suggest that these associations are partially causal. The review synthesizes extensive research on the link between acute and chronic stress and hypercoagulability, outlining a potential pathway from stress to thrombosis risk. Consistent with the allostatic load concept, acute stress-induced hypercoagulability, initially adaptive, can turn maladaptive under chronic stress or excessive acute stress, leading to arterial or venous thrombotic events. Individuals with predisposing factors, including atherosclerosis, thrombophilia, or immobilization, may exhibit an increased risk of thrombotic disease during stress. Contextual sociodemographic characteristics, the stress experience, and coping resources additionally modulate the extent of stress-induced hypercoagulability. Research into the neuroendocrine, cellular, and molecular bases reveals how stress influences platelet activation coagulation and fibrinolysis. The activation of the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis, along with vagal withdrawal, and the effects of catecholamines, cortisol, and vasopressin, are the central mechanisms involved. Hemoconcentration, inflammation, endothelial dysfunction, and thrombopoiesis additionally contribute to stress-induced hypercoagulability. Further research is needed to prove a causal link between chronic stress and hypercoagulability. This includes exploring its implications for the prevention and management of thrombotic diseases in stressed individuals, with a focus on developing effective psychosocial and pharmacological interventions.
Collapse
Affiliation(s)
- Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Chen X, Liu C, Wang J, Du C. Hematopoietic Stem Cells as an Integrative Hub Linking Lifestyle to Cardiovascular Health. Cells 2024; 13:712. [PMID: 38667327 PMCID: PMC11049205 DOI: 10.3390/cells13080712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Despite breakthroughs in modern medical care, the incidence of cardiovascular disease (CVD) is even more prevalent globally. Increasing epidemiologic evidence indicates that emerging cardiovascular risk factors arising from the modern lifestyle, including psychosocial stress, sleep problems, unhealthy diet patterns, physical inactivity/sedentary behavior, alcohol consumption, and tobacco smoking, contribute significantly to this worldwide epidemic, while its underpinning mechanisms are enigmatic. Hematological and immune systems were recently demonstrated to play integrative roles in linking lifestyle to cardiovascular health. In particular, alterations in hematopoietic stem cell (HSC) homeostasis, which is usually characterized by proliferation, expansion, mobilization, megakaryocyte/myeloid-biased differentiation, and/or the pro-inflammatory priming of HSCs, have been shown to be involved in the persistent overproduction of pro-inflammatory myeloid leukocytes and platelets, the cellular protagonists of cardiovascular inflammation and thrombosis, respectively. Furthermore, certain lifestyle factors, such as a healthy diet pattern and physical exercise, have been documented to exert cardiovascular protective effects through promoting quiescence, bone marrow retention, balanced differentiation, and/or the anti-inflammatory priming of HSCs. Here, we review the current understanding of and progression in research on the mechanistic interrelationships among lifestyle, HSC homeostasis, and cardiovascular health. Given that adhering to a healthy lifestyle has become a mainstream primary preventative approach to lowering the cardiovascular burden, unmasking the causal links between lifestyle and cardiovascular health from the perspective of hematopoiesis would open new opportunities to prevent and treat CVD in the present age.
Collapse
Affiliation(s)
| | | | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China; (X.C.); (C.L.)
| | - Changhong Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China; (X.C.); (C.L.)
| |
Collapse
|
8
|
Huang YC, Ko PH, Wu LL. Age-dependent effects of acute stress on the behavior, blood parameters, immunity, and enteric nerves of mice. Behav Brain Res 2024; 461:114848. [PMID: 38185382 DOI: 10.1016/j.bbr.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
The impact of stress on mental and digestive health has been extensively studied, with chronic stress being associated with various disorders. However, age-related differences in the response to acute stress, both behaviorally and physiologically, remain poorly understood. Therefore, this study aimed to develop a model to detect transient stress in mice of different ages. The stressor employed in our experiments was a restraint stress procedure, where mice were subjected to brief periods of immobilization to induce an acute stress response. Male C3H/HeN mice aged 3, 6, 12, and 30 weeks were subjected to acute restrain stress (ARS) by being placed in a 50 ml conical centrifuge tube for 15 min. Subsequently, their behavior, organ tissues, hematological parameters, cortisol concentration, and immune responses were assessed. Following ARS, the increased in time and entries into the center by the 12-week-old mice following stress. In comparison to mice of other ages, those aged 6 weeks demonstrated notable elevations in erythrocytes, platelets, hemoglobin, and hematocrit, all of which were influenced by the time-dependent changes and the recovery process of ARS. Blood corticosterone levels were substantially elevated in all age groups after ARS. Furthermore, ARS induced a notable increase in leukocytes, basophils, residential macrophages, and CD4+ T cells in all age groups except for 3-week-old mice. However, the number of monocyte-derived macrophages and CD8+ T cells did not change significantly. Additionally, mice aged 3 and 6 weeks demonstrated an increase in GFAP+ cells following ARS, whereas NeuN+ cells decreased across all ages. These results suggest that ARS has varying effects on the behavior, cortisol concentration, and quantity of blood cells as well as hepatic immune cells in mice of different ages. These age-dependent responses shed light on the complex interplay between stress and physiological systems and contribute to the broader understanding of stress-related diseases.
Collapse
Affiliation(s)
- Yi-Chen Huang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Pin-Hao Ko
- Department of Traditional Chinese Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 330215, Taiwan
| | - Li-Ling Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Health Innovation Center, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan; Microbiota Research Center, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan.
| |
Collapse
|
9
|
Petrák O, Krátká Z, Holaj R, Zítek M, Nguyen Nikrýnová T, Klímová J, Kološová B, Waldauf P, Michalský D, Novák K, Markvartová A, Zlatohlávek L, Grus T, Dušková J, Widimský J, Zelinka T. Cardiovascular Complications in Pheochromocytoma and Paraganglioma: Does Phenotype Matter? Hypertension 2024; 81:595-603. [PMID: 38152977 DOI: 10.1161/hypertensionaha.123.21902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Adrenaline-producing tumors are mostly characterized by a sudden release of catecholamines with episodic symptoms. Noradrenergic ones are usually less symptomatic and characterized by a continuous overproduction of catecholamines that are released into the bloodstream. Their effects on the cardiovascular system can thus be different. The aim of this study was to determine the prevalence of cardiovascular complications by catecholamine phenotype. METHODS We retrospectively analyzed data on the prevalence of cardiovascular events in 341 consecutive patients with pheochromocytoma and paraganglioma treated from 1995 to 2023. Biochemical catecholamine phenotype was determined based on plasma or urinary catecholamines and metanephrines. RESULTS According to the phenotype, 153 patients had noradrenergic pheochromocytoma and paraganglioma and 188 had adrenergic pheochromocytoma and paraganglioma. In the whole sample, the incidence of serious cardiovascular complications was 28% (95 patients), with no difference between the phenotypes or sexes. The noradrenergic phenotype had significantly more atherosclerotic complications (composite end point of type 1 myocardial infarction and symptomatic peripheral artery disease; odds ratio, 3.58 [95% CI, 1.59-8.83]; P=0.003), while the adrenergic phenotype more often had type 2 myocardial infarction and takotsubo-like cardiomyopathy (OR, 0.24 [95% CI, 0.09-0.57]; P=0.002). These changes remained even after adjustment for conventional risk factors of atherosclerosis. CONCLUSIONS We found a 28% incidence of cardiovascular complications in a consecutive group of patients with pheochromocytoma and paraganglioma. Patients presenting with a noradrenergic phenotype have a higher incidence of atherosclerotic complications, while the adrenergic phenotype is associated with a higher incidence of acute myocardial damage due to takotsubo-like cardiomyopathy.
Collapse
Affiliation(s)
- Ondřej Petrák
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Zuzana Krátká
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Robert Holaj
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Matěj Zítek
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Thi Nguyen Nikrýnová
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Judita Klímová
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Barbora Kološová
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Petr Waldauf
- Department of Anesthesiology, Third Faculty of Medicine, University Hospital Královské Vinohrady, Charles University, Czech Republic (P.W.)
| | - David Michalský
- Department of Urology, 1st Faculty of Medicine (D.M.), Charles University and General University Hospital in Prague, Czech Republic
| | - Květoslav Novák
- 1st Surgical Clinic, Thoracic, Abdominal and Injury Surgery, 1st Faculty of Medicine (K.N.), Charles University and General University Hospital in Prague, Czech Republic
| | - Alice Markvartová
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Lukáš Zlatohlávek
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Tomáš Grus
- 2nd Surgical Clinic, Cardiovascular Surgery, 1st Faculty of Medicine (T.G.), Charles University and General University Hospital in Prague, Czech Republic
| | - Jaroslava Dušková
- Institute of Pathology, 1st Faculty of Medicine (J.D.), Charles University and General University Hospital in Prague, Czech Republic
| | - Jiří Widimský
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| | - Tomáš Zelinka
- 3rd Department of Internal Medicine, Endocrinology and Metabolism, 1st Faculty of Medicine (O.P., Z.K., R.H., M.Z., T.M.P.N.N., J.K., B.K., A.M., L.Z., J.W., T.Z.), Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
10
|
Chen Z, Liu P, Xia X, Cao C, Ding Z, Li X. Low ambient temperature exposure increases the risk of ischemic stroke by promoting platelet activation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169235. [PMID: 38097078 DOI: 10.1016/j.scitotenv.2023.169235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/16/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Accumulating epidemiological evidence suggests the association between low ambient temperature exposure and the risk of ischemic stroke, but the underlying mechanisms remain unclear. OBJECTIVE Given the crucial role of platelet activation and thrombosis in ischemic stroke, this study aims to investigate the effect of ambient temperature on platelet activation through multi-center clinical data in Tianjin as well as animal experiments. METHODS From 2018 to 2020, nearly 3000 ischemic stroke patients from three stroke centers in Tianjin were included in the analysis, among them the ADP induced platelet aggregation rate was available. Meteorological data from the same period had also been collected. After controlling for confounding factors, the generalized additive mixed model (GAMM) was used to evaluate the correlation between environmental temperature and platelet aggregation rate. In further animal experiments, platelet function assessments were conducted on mice from the cold exposure group and the normal temperature group, including platelet aggregation, spreading, and clot retraction. Additionally, tail bleeding and mesentery thrombosis were also tested to monitor hemostasis and thrombosis in vivo. RESULT A nonlinear "S" shaped relationship between outdoor temperature and platelet aggregation was found. Each 1 °C decrease of mean temperature was associated with an increase of 7.77 % (95 % CI: 2.06 % - 13.48 %) in platelet aggregation. The ambient temperature is not related to other platelet parameters. Subgroup analysis found that males, people aged ≥65 years, and hypertensive individuals are more susceptible to temperature changes. Furthermore, animal experiments demonstrated that the increased CIRBP levels and subsequent activation of p-AKT/p-ERK may be one of the reasons for cold exposure induced platelets activation. CONCLUSION Both clinical data and basic research support that low ambient temperature exposure has the potential to increase platelet activation. These results provide a basis for understanding the potential mechanism of temperature variations on the pathogenesis of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhuangzhuang Chen
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Peilin Liu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaoshuang Xia
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Interdisciplinary Innovation Centre for Health and Meteorology, Tianjin, China
| | - Chen Cao
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhongren Ding
- Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin, China; School of Pharmacy, Tianjin Medical University, China.
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China; Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Interdisciplinary Innovation Centre for Health and Meteorology, Tianjin, China.
| |
Collapse
|
11
|
Oh JH, Jun DW. Nonalcoholic fatty liver disease–related extrahepatic complications, associated outcomes, and their treatment considerations. METABOLIC STEATOTIC LIVER DISEASE 2024:101-122. [DOI: 10.1016/b978-0-323-99649-5.00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Liao Y, Chen Z, Yang Y, Shen D, Chai S, Ma Y, Ge R, Wang X, Wang S, Liu S. Antibiotic intervention exacerbated oxidative stress and inflammatory responses in SD rats under hypobaric hypoxia exposure. Free Radic Biol Med 2023; 209:70-83. [PMID: 37806597 DOI: 10.1016/j.freeradbiomed.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
The gut microbiota plays a crucial role in maintaining host nutrition, metabolism, and immune homeostasis, particularly in extreme environmental conditions. However, the regulatory mechanisms of the gut microbiota in animal organisms hypobaric hypoxia exposure require further study. We conducted a research by comparing SD rats treated with an antibiotic (ABX) cocktail and untreated SD rats that were housed in a low-pressure oxygen chamber (simulating low pressure and hypoxic environment at 6000 m altitude) for 30 days. After the experiment, blood, feces, and lung tissues from SD rats were collected for analysis of blood, 16S rRNA amplicon sequencing, and non-targeted metabolomics. The results demonstrated that the antibiotic cocktail-treated SD rats exhibited elevated counts of neutrophil (Neu) and monocyte (Mon) cells, an enrichment of sulfate-reducing bacteria (SBC), reduced levels of glutathione, and accumulated phospholipid compounds. Notably, the accumulation of phospholipid compounds, particularly lysophosphatidic acid (LPA), lipopolysaccharide (LPS), and lysophosphatidylcholine (LPC), along with the aforementioned changes, contributed to heightened oxidative stress and inflammation in the organism. In addition, we explored the resistance mechanisms of SD rats in low-oxygen and low-pressure environments and found that increasing the quantity of the Prevotellaceae and related beneficial bacteria (especially Lactobacillus) could reduce oxidative stress and inflammation. These findings offer valuable insights into enhancing the adaptability of low-altitude animals under hypobaric hypoxia exposure.
Collapse
Affiliation(s)
- Yang Liao
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Zheng Chen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yingkui Yang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Di Shen
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shatuo Chai
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Yan Ma
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, China
| | - Xun Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China
| | - Shuxiang Wang
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| | - Shujie Liu
- College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining, 810016, China.
| |
Collapse
|
13
|
Madias JE. Delving in the follow-up hemostatic changes in patients with Takotsubo syndrome. Int J Cardiol 2023; 391:131307. [PMID: 37652273 DOI: 10.1016/j.ijcard.2023.131307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023]
Affiliation(s)
- John E Madias
- Icahn School of Medicine at Mount Sinai, New York, NY, and the Division of Cardiology, Elmhurst Hospital Center, Elmhurst, NY, United States of America.
| |
Collapse
|
14
|
Hernández-Barrientos D, Pelayo R, Mayani H. The hematopoietic microenvironment: a network of niches for the development of all blood cell lineages. J Leukoc Biol 2023; 114:404-420. [PMID: 37386890 DOI: 10.1093/jleuko/qiad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/25/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Blood cell formation (hematopoiesis) takes place mainly in the bone marrow, within the hematopoietic microenvironment, composed of a number of different cell types and their molecular products that together shape spatially organized and highly specialized microstructures called hematopoietic niches. From the earliest developmental stages and throughout the myeloid and lymphoid lineage differentiation pathways, hematopoietic niches play a crucial role in the preservation of cellular integrity and the regulation of proliferation and differentiation rates. Current evidence suggests that each blood cell lineage develops under specific, discrete niches that support committed progenitor and precursor cells and potentially cooperate with transcriptional programs determining the gradual lineage commitment and specification. This review aims to discuss recent advances on the cellular identity and structural organization of lymphoid, granulocytic, monocytic, megakaryocytic, and erythroid niches throughout the hematopoietic microenvironment and the mechanisms by which they interconnect and regulate viability, maintenance, maturation, and function of the developing blood cells.
Collapse
Affiliation(s)
- Daniel Hernández-Barrientos
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| | - Rosana Pelayo
- Onco-Immunology Laboratory, Eastern Biomedical Research Center, IMSS, Km 4.5 Atlixco-Metepec, 74360, Puebla, Mexico
| | - Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| |
Collapse
|
15
|
Teixeira IG, Verzola MR, Filipini RE, Speretta GF. The effects of a firefighting simulation on the vascular and autonomic functions and cognitive performance: a randomized crossover study. Front Physiol 2023; 14:1215006. [PMID: 37811491 PMCID: PMC10551144 DOI: 10.3389/fphys.2023.1215006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction: During firefighting, physical and cognitive demands increase. However, the stress inherent to these events can decrease cognitive performance and increase the risk of cardiovascular events in firefighters. Thus, this crossover study aimed to evaluate the effects of a firefighting Simulation on cognitive performance and vascular and autonomic functions in military firefighters. Methods: Sixteen firefighters (37.8 ± 5.6 years) underwent anthropometry, mental health status, and sleep quality assessments. They randomly performed two interventions, Simulation (Firefighting tasks; 10.0 ± 1.1 min) and Control (rest for 10 min), on different days. After both interventions, cognitive performance was assessed using the Stroop Test, Paced Auditory Serial Addition Test, and Trail Making Test. Then, the vascular function was assessed using ultrasonography through the carotid artery reactivity to the cold pressor test. The arterial pressure, heart rate, and cardiac intervals were recorded before interventions. The cardiac intervals were also measured during the cold pressor test. Student's t-test and Wilcoxon were used for comparisons between Control and Simulation and the analysis of variance for repeated measures was used for comparison over time during the cold pressor test. A significance level of p < 0.05 was adopted. Results: Although the mean and maximum heart rate were higher before the Simulation (p < 0.0001), all the heart rate variability parameters (p > 0.05) and mean arterial pressure (p > 0.3795) were similar before the interventions. After Simulation, the cognitive performance was similar to Control (p > 0.05), except for the improvement in Stroop Test part B (p < 0.0001). After Simulation, carotid artery reactivity was attenuated (p < 0.0010). During the cold pressor test, the high-frequency band of the heart rate variability was lower after the Simulation (p < 0.0104). Discussion: Although firefighting Simulation did not substantially change cognitive performance, the lower carotid artery reactivity and parasympathetic modulation to the heart during the cold pressor test may contribute to greater vulnerability to cardiovascular events in firefighters on duty.
Collapse
Affiliation(s)
- Iara G. Teixeira
- Post-Graduate Program in Neurosciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, Biological Sciences Centre, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Marcio R. Verzola
- Military Firefighters Corps of Santa Catarina, Florianópolis, Brazil
| | - Richard E. Filipini
- Department of Physiological Sciences, Biological Sciences Centre, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Guilherme F. Speretta
- Post-Graduate Program in Neurosciences, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, Biological Sciences Centre, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
16
|
Zhang GC, Wu YJ, Liu FQ, Chen Q, Sun XY, Qu QY, Fu HX, Huang XJ, Zhang XH. β2-adrenergic receptor agonist corrects immune thrombocytopenia by reestablishing the homeostasis of T cell differentiation. J Thromb Haemost 2023; 21:1920-1933. [PMID: 36972787 DOI: 10.1016/j.jtha.2023.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND The sympathetic nerve is known to regulate immune responses in autoimmunity. Aberrant T cell immunity plays a vital role in immune thrombocytopenia (ITP) pathogenesis. The spleen is the primary site of platelet destruction. However, little is known whether and how splenic sympathetic innervation and neuroimmune modulation contribute to ITP pathogenesis. OBJECTIVES To determine the sympathetic distribution in the spleen of ITP mice and the association between splenic sympathetic nerves and T cell immunity in ITP development, and to evaluate the treatment potential of β2-adrenergic receptor (β2-AR) in ITP. METHODS Chemical sympathectomy was performed in an ITP mouse model with 6-hydroxydopamine and treated with β2-AR agonists to evaluate the effects of sympathetic denervation and activation. RESULTS Decreased sympathetic innervation in the spleen of ITP mice was observed. Significantly increased percentages of Th1 and Tc1 cells and reduced percentages of regulatory T cells (Tregs) were also observed in ITP mice with chemical sympathectomy (ITP-syx mice) relative to mice without sympathectomy (controls). Expression of genes associated with Th1, including IFN-γ and IRF8, was significantly upregulated, whereas genes associated with Tregs, including Foxp3 and CTLA4, were significantly downregulated in ITP-syx mice compared with controls. Furthermore, β2-AR restored the percentage of Tregs and increased platelet counts at days 7 and 14 in ITP mice. CONCLUSION Our findings indicate that decreased sympathetic distribution contributes to ITP pathogenesis by disturbing the homeostasis of T cells and that β2-AR agonists have potential as a novel treatment for ITP.
Collapse
Affiliation(s)
- Gao-Chao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ye-Jun Wu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Feng-Qi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qi Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xue-Yan Sun
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qing-Yuan Qu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; National Clinical Research Center for Hematologic Disease, Beijing, China.
| |
Collapse
|
17
|
Hsu YT, Chen LH, Liu YH, Chu SK, Chen TY, Tsai KJ, Shen MR, Liu W. Electrical Sympathetic Neuromodulation Protects Bone Marrow Niche and Drives Hematopoietic Regeneration during Chemotherapy. SMALL METHODS 2023; 7:e2201300. [PMID: 36843214 DOI: 10.1002/smtd.202201300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/24/2023] [Indexed: 06/09/2023]
Abstract
The sympathetic nervous system (SNS) of the bone marrow regulates the regeneration and mobilization of hematopoietic stem cells. Chemotherapy can damage bone marrow SNS, which impairs hematopoietic regeneration and aggravates hematologic toxicities. This leads to long-term bone marrow niche damage and increases mortality in patients undergoing chemotherapy. Electrical neuromodulation has been used to improve functional recovery after peripheral nerve injury. This study demonstrates that electrical sympathetic neuromodulation (ESN) of bone marrow can protect the bone marrow niche from chemotherapy-induced injury. Using carboplatin-treated rats, the SNS via the sciatic nerve innervating the femoral marrow with the effective protocol for bone marrow sympathetic activation is electrically stimulated. ESN can mediate several hematopoietic stem cells maintenance factors and promote hematopoietic regeneration after chemotherapy. It also activates adrenergic signals and reduces the release of pro-inflammatory cytokines, particularly interleukin-1 β, which contribute to chemotherapy-related nerve injury. Consequently, the severity of chemotherapy-related leukopenia, thrombocytopenia, and mortality can be reduced by ESN. As a result, in contrast to current drug-based treatment, such as granulocyte colony-stimulating factor, ESN can be a disruptive adjuvant treatment by protecting and modulating bone marrow function to reduce hematologic toxicity during chemotherapy.
Collapse
Affiliation(s)
- Ya-Ting Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701401, Taiwan
- Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704302, Taiwan
| | - Li-Hsien Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701401, Taiwan
| | - Ya-Hui Liu
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704302, Taiwan
| | - Shih-Kai Chu
- Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, 704302, Taiwan
| | - Tsai-Yun Chen
- Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704302, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701401, Taiwan
- Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine National Cheng Kung University, Tainan, 704302, Taiwan
| | - Meng-Ru Shen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701401, Taiwan
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 701401, Taiwan
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704302, Taiwan
| | - Wentai Liu
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Electrical and Computer Engineering, University of California, Los Angeles, CA, 90095, USA
- Brain Research Institute, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
18
|
Huang L, Xu J, Zhang H, Wang M, Zhang Y, Lin Q. Application and investigation of thrombopoiesis-stimulating agents in the treatment of thrombocytopenia. Ther Adv Hematol 2023; 14:20406207231152746. [PMID: 36865986 PMCID: PMC9972067 DOI: 10.1177/20406207231152746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/06/2023] [Indexed: 03/02/2023] Open
Abstract
Platelets, derived from a certain subpopulation of megakaryocytes, are closely related to hemostasis, coagulation, metastasis, inflammation, and cancer progression. Thrombopoiesis is a dynamic process regulated by various signaling pathways in which thrombopoietin (THPO)-MPL is dominant. Thrombopoiesis-stimulating agents could promote platelet production, showing therapeutic effects in different kinds of thrombocytopenia. Some thrombopoiesis-stimulating agents are currently used in clinical practices to treat thrombocytopenia. The others are not in clinical investigations to deal with thrombocytopenia but have potential in thrombopoiesis. Their potential values in thrombocytopenia treatment should be highly regarded. Novel drug screening models and drug repurposing research have found many new agents and yielded promising outcomes in preclinical or clinical studies. This review will briefly introduce thrombopoiesis-stimulating agents currently or potentially valuable in thrombocytopenia treatment and summarize the possible mechanisms and therapeutic effects, which may enrich the pharmacological armamentarium for the medical treatment of thrombocytopenia.
Collapse
Affiliation(s)
- Lejun Huang
- Division of Cell, Developmental and Integrative
Biology, School of Medicine, South China University of Technology,
Guangzhou, P.R. China
| | - Jianxuan Xu
- Division of Cell, Developmental and Integrative
Biology, School of Medicine, South China University of Technology,
Guangzhou, P.R. China
| | - Huaying Zhang
- Division of Cell, Developmental and Integrative
Biology, School of Medicine, South China University of Technology,
Guangzhou, P.R. China
| | - Mengfan Wang
- Division of Cell, Developmental and Integrative
Biology, School of Medicine, South China University of Technology,
Guangzhou, P.R. China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative
Biology, School of Medicine, South China University of Technology,
Guangzhou, P.R. China
| | | |
Collapse
|
19
|
Akt-mediated mitochondrial metabolism regulates proplatelet formation and platelet shedding post vasopressin exposure. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:344-358. [PMID: 36700501 DOI: 10.1016/j.jtha.2022.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Platelet shedding from mature megakaryocytes (MKs) in thrombopoiesis is the critical step for elevating circulating platelets fast and efficiently, however, the underlying mechanism is still not well-illustrated, and the therapeutic targets and candidates are even less. OBJECTIVES In order to investigate the mechanisms for platelet shedding after vasopressin treatment and find new therapeutic targets for thrombocytopenia. METHODS Platelet production was evaluated both in vivo and in vitro after arginine vasopressin (AVP) administration. The underlying biological mechanism of AVP-triggered thrombopoiesis were then investigated by a series of molecular and bioinformatics techniques. RESULTS it is observed that proplatelet formation and platelet shedding in the final stages of thrombopoiesis promoted by AVP, an endogenous hormone, can quickly increases peripheral platelets. This rapid elevation is thus able to speed up platelet recovery after radiation as expected. The mechanism analysis reveal that proplatelet formation and platelet release from mature MKs facilitated by AVP is mainly mediated by Akt-regulated mitochondrial metabolism. In particular, phosphorylated Akt regulates mitochondrial metabolism through driving the association of hexokinase-2 with mitochondrial voltage dependent anion channel-1 in AVP-mediated thrombopoiesis. Further studies suggest that this interaction is stabilized by IκBα, the expression of which is controlled by insulin-regulated membrane aminopeptidase. CONCLUSION these data demonstrate that phosphorylated Akt-mediated mitochondrial metabolism regulates platelet shedding from MKs in response to AVP, which will provide new therapeutic targets and further drug discovery clues for thrombocytopenia treatment.
Collapse
|
20
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
21
|
Petzold T, Zhang Z, Ballesteros I, Saleh I, Polzin A, Thienel M, Liu L, Ul Ain Q, Ehreiser V, Weber C, Kilani B, Mertsch P, Götschke J, Cremer S, Fu W, Lorenz M, Ishikawa-Ankerhold H, Raatz E, El-Nemr S, Görlach A, Marhuenda E, Stark K, Pircher J, Stegner D, Gieger C, Schmidt-Supprian M, Gaertner F, Almendros I, Kelm M, Schulz C, Hidalgo A, Massberg S. Neutrophil "plucking" on megakaryocytes drives platelet production and boosts cardiovascular disease. Immunity 2022; 55:2285-2299.e7. [PMID: 36272416 PMCID: PMC9767676 DOI: 10.1016/j.immuni.2022.10.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/23/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
Abstract
Intravascular neutrophils and platelets collaborate in maintaining host integrity, but their interaction can also trigger thrombotic complications. We report here that cooperation between neutrophil and platelet lineages extends to the earliest stages of platelet formation by megakaryocytes in the bone marrow. Using intravital microscopy, we show that neutrophils "plucked" intravascular megakaryocyte extensions, termed proplatelets, to control platelet production. Following CXCR4-CXCL12-dependent migration towards perisinusoidal megakaryocytes, plucking neutrophils actively pulled on proplatelets and triggered myosin light chain and extracellular-signal-regulated kinase activation through reactive oxygen species. By these mechanisms, neutrophils accelerate proplatelet growth and facilitate continuous release of platelets in steady state. Following myocardial infarction, plucking neutrophils drove excessive release of young, reticulated platelets and boosted the risk of recurrent ischemia. Ablation of neutrophil plucking normalized thrombopoiesis and reduced recurrent thrombosis after myocardial infarction and thrombus burden in venous thrombosis. We establish neutrophil plucking as a target to reduce thromboischemic events.
Collapse
Affiliation(s)
- Tobias Petzold
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Corresponding author
| | - Zhe Zhang
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Iván Ballesteros
- Program of Cardiovascular Regeneration, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Inas Saleh
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty of the Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Manuela Thienel
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Lulu Liu
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Qurrat Ul Ain
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Vincent Ehreiser
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Christian Weber
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Badr Kilani
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Pontus Mertsch
- Medizinische Klinik und Poliklinik V, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Jeremias Götschke
- Medizinische Klinik und Poliklinik V, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Sophie Cremer
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty of the Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Wenwen Fu
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Hellen Ishikawa-Ankerhold
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Elisabeth Raatz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Shaza El-Nemr
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University of Munich, 80636 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany
| | - Esther Marhuenda
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Joachim Pircher
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - David Stegner
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, 97070 Würzburg, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany,Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, 80333 Munich, Germany,Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69117 Heidelberg, Germany
| | - Florian Gaertner
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Isaac Almendros
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain,CIBER de Enfermedades Respiratorias, 28029 Madrid, Spain
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty of the Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany
| | - Andrés Hidalgo
- Program of Cardiovascular Regeneration, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain,Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany,Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians- University Munich, 81377 Munich, Germany,Corresponding author
| |
Collapse
|
22
|
Oh T, Ogawa K, Nagoshi T, Minai K, Ogawa T, Kawai M, Yoshimura M. Relationship between haemodynamic indicators and haemogram in patients with heart failure. ESC Heart Fail 2022; 10:955-964. [PMID: 36478404 PMCID: PMC10053360 DOI: 10.1002/ehf2.14258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/08/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Pulmonary congestion, reduced cardiac output, neurohumoral factor activation, and decreased renal function associated with decreased cardiac function may have various effects on haemograms. The relationship between these factors and haemograms in patients with heart failure has not been sufficiently investigated. Recently, it was suggested that the lungs are an important site for platelet (Plt) biosynthesis and that it is necessary to study the relationship between pulmonary congestion and Plt count in heart failure in detail. In this study, we examined the relationship between various haemodynamic indicators and haemograms in detail using statistical analyses. METHODS AND RESULTS A total of 345 patients who underwent cardiac catheterization for the evaluation of cardiac function between 1 January 2015 and 31 December 2020 were included in the study. Haemodynamic indices, including left ventricular end-diastolic pressure (LVEDP) and cardiac index (CI), were measured. Plasma noradrenaline (Nor) concentration, estimated glomerular filtration rate (eGFR), white blood cell (WBC) count, haemoglobin (Hb) level, and Plt count were measured using blood samples collected at the same time. Structural equation modelling (SEM) was used to examine the relationship between LVEDP, CI, plasma Nor concentration, eGFR, WBC count, Hb level, and Plt count. Bayesian inference using SEM was performed for Plt count. A total of 345 patients (mean age: 66.0 ± 13.2 years) were included in this study, and 251 (73%) patients were men. After simple and multiple regression analyses, path diagrams were drawn and analysed using SEM. LVEDP showed a significant negative relationship with Plt count (standardized estimate: -0.129, P = 0.015), and CI showed a significant negative relationship with Hb level (standardized estimate: -0.263, P < 0.001). Plasma Nor concentration showed a significant positive relationship with WBC count (standardized estimate: 0.165, P = 0.003) and Plt count (standardized estimate: 0.198, P < 0.001). The eGFR had a significant positive relationship with Hb level (standardized estimate: 0.274, P < 0.001). Bayesian inference using SEM revealed no relationship between LVEDP and Hb level or WBC count but a significant negative relationship between LVEDP and Plt count. CONCLUSIONS LVEDP, CI, plasma Nor concentration, and eGFR were related to WBC count, Hb level, and Plt count in patients with heart failure. There was a strong relationship between elevated LVEDP and decreased Plt count, suggesting that pressure overload on the lungs may interfere with the function of the lung as a site of Plt biosynthesis.
Collapse
Affiliation(s)
- Takuya Oh
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Kosuke Minai
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Takayuki Ogawa
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| |
Collapse
|
23
|
Airapetov MI, Eresko SO, Bychkov ER, Lebedev AA, Shabanov PD. Effect of Ethanol on Platelet Biology. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2022. [DOI: 10.1134/s1990750822040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
24
|
Wang Y, Qu M, Qiu Z, Zhu S, Chen W, Guo K, Miao C, Zhang H. Surgical Stress and Cancer Progression: New Findings and Future Perspectives. Curr Oncol Rep 2022; 24:1501-1511. [PMID: 35763189 DOI: 10.1007/s11912-022-01298-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW The stress response to surgery is essential for maintaining homeostasis and exhibits anti-tumor effects; however, an ongoing and exaggerated stress response may have adverse clinical consequences and even promote cancer progression. This review will discuss the complex relationship between surgical stress and cancer progression. RECENT FINDINGS Surgical stress exhibits both anti-tumor and cancer-promoting effects by causing changes in the neuroendocrine, circulatory, and immune systems. Many studies have found that many mechanisms are involved in the process, and the corresponding targets could be applied for cancer therapy. Although surgical stress may have anti-tumor effects, it is necessary to inhibit an excessive stress response, mostly showing cancer-promoting effects.
Collapse
Affiliation(s)
- Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhiyun Qiu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
- Department of Anesthesiology, Fudan University Jinshan Hospital, Shanghai, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
25
|
Zhu J, Duan Y, Duo D, Yang J, Bai X, Liu G, Wang Q, Wang X, Qu N, Zhou Y, Li X. High-altitude Hypoxia Influences the Activities of the Drug-Metabolizing Enzyme CYP3A1 and the Pharmacokinetics of Four Cardiovascular System Drugs. Pharmaceuticals (Basel) 2022; 15:ph15101303. [PMID: 36297415 PMCID: PMC9612038 DOI: 10.3390/ph15101303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: High-altitude hypoxia has been shown to affect the pharmacokinetic properties of drugs. Although there is a high incidence of cardiovascular disease among individuals living in high-altitude areas, studies on the effect of high-altitude hypoxia on the pharmacokinetic properties of cardiovascular drugs are limited. (2) Methods: The aim of this study was to evaluate the pharmacokinetics of nifedipine, bosentan, simvastatin, sildenafil, and their respective main metabolites, dehydronifedipine, hydroxybosentan, simvastatin hydroxy acid, and N-desmethyl sildenafil, in rats exposed to high-altitude hypoxia. Additionally, the protein and mRNA expression of cytochrome P450 3A1 (CYP3A1), a drug-metabolizing enzyme, were examined. (3) Results: There were significant changes in the pharmacokinetic properties of the drugs in rats exposed to high-altitude hypoxia, as evidenced by an increase in the area under the curve (AUC) and the half-life (t1/2z) and a decrease in total plasma clearance (CLz/F). However, most of these changes were reversed when the rats returned to a normoxic environment. Additionally, there was a significant decrease in CYP3A1 expression in rats exposed to high-altitude hypoxia at both the protein and mRNA levels. (4) Conclusions: High-altitude hypoxia suppressed the metabolism of the drugs, indicating that the pharmacokinetics of the drugs should be re-examined, and the optimal dose should be reassessed in patients living in high-altitude areas.
Collapse
Affiliation(s)
- Junbo Zhu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810000, China
| | - Yabin Duan
- Department of Clinical Pharmacy, Qinghai University Affiliated Hospital, Xining 810000, China
| | - Delong Duo
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
| | - Jianxin Yang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
| | - Xue Bai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
| | - Guiqin Liu
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
| | - Qian Wang
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
| | - Xuejun Wang
- Department of Anesthesiology, Red Cross Hospital of Qinghai, Xining 810000, China
| | - Ning Qu
- Department of Anesthesiology, Qinghai Hospital of Traditional Chinese Medicine, Xining 810000, China
| | - Yang Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
- Correspondence: (Y.Z.); (X.L.)
| | - Xiangyang Li
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810000, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810000, China
- Correspondence: (Y.Z.); (X.L.)
| |
Collapse
|
26
|
Chen Z, Liu P, Xia X, Wang L, Li X. The underlying mechanisms of cold exposure-induced ischemic stroke. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 834:155514. [PMID: 35472344 DOI: 10.1016/j.scitotenv.2022.155514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
Growing evidence suggests that cold exposure is to some extent a potential risk factor for ischemic stroke. At present, although the mechanism by which cold exposure induces ischemic stroke is not fully understood, some potential mechanisms have been mentioned. First, the seasonal and temperature variability of cerebrovascular risk factors (hypertension, hyperglycemia, hyperlipidemia, atrial fibrillation) may be involved. Moreover, the activation of sympathetic nervous system and renin-angiotensin system and their downstream signaling pathways (pro-inflammatory AngII, activated platelets, and dysfunctional immune cells) are also major contributors. Finally, the influenza epidemics induced by cold weather are also influencing factors that cannot be ignored. This article is the first to systematically and comprehensively describe the underlying mechanism of cold-induced ischemic stroke, aiming to provide more preventive measures and medication guidance for stroke-susceptible individuals in cold season, and also provide support for the formulation of public health policies.
Collapse
Affiliation(s)
- Zhuangzhuang Chen
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Peilin Liu
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiaoshuang Xia
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Lin Wang
- Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
27
|
Airapetov MI, Eresko SO, Bychkov ER, Lebedev AA, Shabanov PD. [Effect of ethanol on platelet biology]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:81-92. [PMID: 35485482 DOI: 10.18097/pbmc20226802081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In recent years, interest in the study of platelets, significantly increased due to recent discoveries providing convincing evidence that their functions by are not limited to their participation in the blood coagulation mechanism. Many works are devoted to the study of the functional state of platelets under conditions of acute and chronic alcohol exposure. The results of such studies can be useful for the development of new markers of the degree of alcohol intoxication of the body for the subsequent choice of the method drug correction of disorders caused by acute or chronic alcohol effects. The review summarizes results in vivo and in vitro of studies performed during more than 60 years on the effect of ethanol on the biogenesis, number, morphology and biochemistry of platelets.
Collapse
Affiliation(s)
- M I Airapetov
- Department of Neuropharmacology, Institute of Experimental Medicine, Saint Petersburg, Russia; Department of Pharmacology, St. Petersburg State Pediatric Medical University, Saint Petersburg, Russia
| | - S O Eresko
- Department of Neuropharmacology, Institute of Experimental Medicine, Saint Petersburg, Russia; Research and Training Center of Molecular and Cellular Technologies, Saint Petersburg, Russia
| | - E R Bychkov
- Department of Neuropharmacology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - A A Lebedev
- Department of Neuropharmacology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - P D Shabanov
- Department of Neuropharmacology, Institute of Experimental Medicine, Saint Petersburg, Russia; Department of Pharmacology, Kirov Military Medical Academy, Saint Petersburg, Russia
| |
Collapse
|
28
|
Zhao L, Meng X, Mei Q, Fan H, Liu Y, Zhou X, Zhu H, Zhang S. Risk Factors for Cardiac Complications in Patients With Pheochromocytoma and Paraganglioma: A Retrospective Single-Center Study. Front Endocrinol (Lausanne) 2022; 13:877341. [PMID: 35721724 PMCID: PMC9199364 DOI: 10.3389/fendo.2022.877341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Catecholamine excess arising from pheochromocytomas and paragangliomas (PPGLs) can cause a wide spectrum of cardiac manifestations. Although there are reviews of reported cases, these reviews lack detailed data, which makes it impossible to perform an accurate analysis. In this study, we conducted a comprehensive analysis of cardiovascular complications (CCs), including PPGL-related myocardial injury, cardiogenic shock, and arrhythmias requiring antiarrhythmic therapy, in a large cohort of patients with PPGL. METHODS We retrospectively analyzed the clinical data of consecutive patients with PPGL admitted between January 2018 and June 2020. The prevalence and the characteristics of patients with CCs were investigated. Moreover, comparisons were made between patients with and without CCs. RESULTS Compared with the non-CC group, the percentage of men was significantly lower (14/41 vs.92/175, 34.1% vs. 52.6%, p = 0.034) and the proportion of patients with paroxysmal hypertension was significantly higher (13/41 vs.29/173, 31.7% vs.16.8%, p = 0.03) in the CC group. More patients showed excessive sweating (19/41 vs 64/175, 46.3% vs. 24.0%, p = 0.004) and PPGL crisis (7/41 vs. 10/175, 17.1% vs.5.7%, p=0.035) in the CC group. In terms of laboratory findings, higher white blood cell [7.36 (6.49, 20.23) vs. 5.95 (5.1, 6.97)×109/L, p<0.001] and platelet [339.28 ± 108.54 vs. 250.66 ± 70.83(×109/L), p = 0.021] counts were more common in the CC group. There was also a higher prevalence of combination-producing PPGL in the CC group (13/24 vs.20/149, 54.2% vs.13.4%, p<0.001). However, the tumor size, invasive behavior on histology, and hemorrhage or necrosis on histology did not differ between the two groups. Platelet count [odds ratio (OR): 1.009; 95% confidence interval (CI) 1.001-1.016; p=0.023] and combination-secreting PPGL (OR: 5.009; 95% CI 1.365-18.38; p=0.015) are independent risk factors for CCs in patients with PPGL. CONCLUSIONS In patients with PPGL, even in the absence of signs and symptoms of CCs, a work up of cardiology should be strongly considered. Importantly, if patients with PPGLs have higher platelet counts and the combination-secreting pattern, they are more likely to have CCs. Thus, a careful cardiac evaluation should be performed.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Meng
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - QiMin Mei
- Department of Emergency, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hua Fan
- Department of Urology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - YeCheng Liu
- Department of Emergency, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: YeCheng Liu, ; XianLiang Zhou, ; HuaDong Zhu, ; ShuYang Zhang,
| | - XianLiang Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: YeCheng Liu, ; XianLiang Zhou, ; HuaDong Zhu, ; ShuYang Zhang,
| | - HuaDong Zhu
- Department of Emergency, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: YeCheng Liu, ; XianLiang Zhou, ; HuaDong Zhu, ; ShuYang Zhang,
| | - ShuYang Zhang
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: YeCheng Liu, ; XianLiang Zhou, ; HuaDong Zhu, ; ShuYang Zhang,
| |
Collapse
|
29
|
Sandrini L, Amadio P, Ieraci A, Malara A, Werba JP, Soprano PM, Balduini A, Zarà M, Bonomi A, Veglia F, Colombo GI, Popoli M, Lee FS, Tremoli E, Barbieri SS. The α 2-adrenergic receptor pathway modulating depression influences the risk of arterial thrombosis associated with BDNFVal66Met polymorphism. Biomed Pharmacother 2021; 146:112557. [PMID: 34965503 DOI: 10.1016/j.biopha.2021.112557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is associated with thrombotic risk and arterial events, its proper management is strongly recommended in coronary artery disease (CAD) patients. We have previously shown that the Brain-Derived Neurotrophic Factor (BDNF)Val66Met polymorphism, related to depression, is associated with arterial thrombosis in mice, and with an increased risk of acute myocardial infarction in humans. Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show that desipramine, a norepinephrine reuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrow megakaryocyte number and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. The in vitro data confirm the enhanced procoagulant activity and the alpha2A-adrenergic receptor (α2A-ADR) overexpression found in BDNFMet/Met mice and we provide evidence that, in presence of Met variant, norepinephrine is crucial to up-regulate procoagulant activity and to enhance platelet generation. The α2-ADR antagonist rauwolscine rescues the prothrombotic phenotype in BDNFMet/Met mice and reduces procoagulant activity and platelet generation in cells transfected with BDNFMet plasmid or exposed to pro-BDNFMet peptide. Finally, we show that homozygous BDNFMet/Met CAD patients have hyper-reactive platelets overexpressing abundant α2A-ADR. The great proplatelet release from their megakaryocytes well reflects their higher circulating platelet number compared to BDNFVal/Val patients. These data reveal an unprecedented described role of Met allele in the dysregulation of norepinephrine/α2A-ADR pathway that may explain the predisposition to arterial thrombosis. Overall, the development of α2A-ADR inhibitors might represent a pharmacological treatment for depression-associated thrombotic conditions in this specific subgroup of CAD patients.
Collapse
Affiliation(s)
| | | | - Alessandro Ieraci
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - José P Werba
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - Marta Zarà
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Alice Bonomi
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, USA
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Maria Cecilia Hospital, Cotignola, Italy
| | | |
Collapse
|
30
|
Mo C, Wang Y, Yue Z, Hu D, Yin C. Influence of exercise test on platelet function in patients with coronary arterial disease: A systematic review. Medicine (Baltimore) 2021; 100:e24932. [PMID: 33663130 PMCID: PMC7909175 DOI: 10.1097/md.0000000000024932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/29/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Exercise test (ET) may have adverse effects on platelet function and induce acute thrombotic events in patients with coronary artery disease (CAD). The aim of this study is to investigate the platelet function and evaluate the risk of thrombotic events in CAD patients during ET. METHODS Pubmed, Embase, Cochrane Library, and Web of Science were searched for a systematic review from initiation to October 2019. The inclusion criteria were controlled clinical trails as study design; investigating platelet function in CAD patients during ET; with ET carried out by treadmill or bicycle ergometer; written in English. Included articles were screened based on title/abstract and full-text review by 2 independent reviewers. Platelet aggregation (PA), platelet surface expression of CD62p and PAC-1, plasma levels of platelet factor 4 (PF4) and beta-thromboglobulin (β-TG) were evaluated before and after ET. RESULTS Eighteen articles were included out of the 427 references initially identified. In most of the studies included ET was terminated because of limited symptoms. Prior to ET, no difference in platelet aggregation was observed in CAD patients compared with healthy controls in majority of the studies, with or without the treatment with Aspirin. Dual anti-platelet therapy suppressed adenosine diphosphate (ADP)-induced platelet aggregation at rest. After ET, platelet aggregation, the serum levels of β-thromboglobulin were found unchanged in majority of studies and platelet factor-4 were found unchanged in half of studies. The expression of platelet surface markers were elevated by ET in a few study. CONCLUSION Symptom-limited exercise test did not affect platelet function in patients with coronary artery disease; however exercise to higher intensity may induce platelet activation.
Collapse
Affiliation(s)
- Chunhua Mo
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing
| | - Yanhui Wang
- Cardiac Rehabilitation Center, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing
| | - Zong Yue
- Cardiac Rehabilitation Center, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing
| | - Dayi Hu
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing
| | - Chun Yin
- Department of Cardiology, First Affiliated Hospital of Chongqing Medical University, Chongqing
- Department of Cardiology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| |
Collapse
|
31
|
Gonzalez-Villalva A, Bizarro-Nevares P, Rojas-Lemus M, Ustarroz-Cano M, López-Valdez N, García-Peláez I, Albarrán-Alonso JC, Barbosa-Barrón F, Fortoul TI. A brief review of the biology of megakaryocytes and platelets and their role in thrombosis associated with particulate air pollution. Toxicol Ind Health 2021; 37:164-172. [PMID: 33506746 DOI: 10.1177/0748233720986352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Air pollution is a worldwide public health issue and it is associated with millions of premature deaths due to cancer, thrombosis, and pulmonary and cardiovascular diseases. Thrombosis is the excessive clotting that blocks a blood vessel, and its etiology is multifactorial. In recent years, growing evidence has linked air pollution, especially particulate matter (PM) and metals, to the development of thrombosis. PM and metals induce lung and systemic inflammation and oxidative stress that are frequent mechanisms in thrombosis. Platelets are important effectors of physiological hemostasis and pathological thrombosis. They are responsible for the formation of the initial plug and are important in the cellular model of coagulation. Therefore, any changes in their morphology or function or an increase in activation could be extremely relevant in thrombosis. Megakaryocytes (MKs) in the bone marrow and in the lungs are the precursor cells of platelets, and the latter is the first organ injured by air pollution. There is substantial evidence of the effect that PM and metals have on platelets, but there is almost no research about the effect of PM and metals on MKs. It is very likely that the alterations produced by air pollution originate in these cells. In this article, we review the biology of MKs and platelets and their role in particulate air pollution-related thrombosis to emphasize the need for further research in this field.
Collapse
Affiliation(s)
- Adriana Gonzalez-Villalva
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Patricia Bizarro-Nevares
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Marcela Rojas-Lemus
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Ustarroz-Cano
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Nelly López-Valdez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Isabel García-Peláez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Juan Carlos Albarrán-Alonso
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Fernanda Barbosa-Barrón
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Teresa I Fortoul
- Departamento de Biología Celular y Tisular, Facultad de Medicina, 7180Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
32
|
Acute platelet response to aneurysmal subarachnoid hemorrhage depends on severity and distribution of bleeding: an observational cohort study. Neurosurg Rev 2020; 44:2647-2658. [PMID: 33241455 DOI: 10.1007/s10143-020-01444-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/21/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Microthrombosis after aneurysmal subarachnoid hemorrhage (aSAH) is considered to initiate neuroinflammation, vessel remodeling, and blood-brain barrier leakage. We aimed to verify the hypothesis that the intensity of thrombogenicity immediately after aSAH depends on the amount and distribution of extravasated blood. This observational cohort study included 37 consecutive aSAH patients admitted no longer than 24 h after ictus. Volumes of subarachnoid and intraventricular hemorrhages as well as the Subarachnoid Hemorrhage Early Brain Edema Scale (SEBES) score were calculated in each case. Platelet system status was described by platelet count (PLT), mean platelet volume (MPV), MPV to PLT ratio, and platelet-large cell ratio (P-LCR). Median hemorrhage volume amounted to 11.4 ml (interquartile range 2.8-26.8 ml). Patients with more severe hemorrhage had lower PLT and higher MPV to PLT ratio (ρ = - 0.49, p < .002; ρ = 0.50, p < .002, respectively). PLT decreased by 2.80 G/l per 1 ml of hemorrhage volume (95% CL 1.30-4.30, p < .001). Further analysis revealed that intraventricular hemorrhage volume was associated with P-LCR and MPV (ρ = 0.34, p < .039; ρ = 0.33, p < .048, respectively), whereas SAH volume with PLT and MPV:PLT ratio (ρ = - 0.40, p < .013; ρ = 0.41, p < .013, respectively). The odds of unfavorable neurological outcome increased 3.95 times per 1 fl of MPV (95% CI 1.19-13.12, p < .025). MPV was independently correlated with SEBES (ρ = 0.44, p < .006). This study demonstrated that the extent and distribution of aneurysmal subarachnoid hemorrhage are related to different types of acute platelet response, which may be interpreted as local and systemic thrombogenicity. Increased mean platelet volume measured in the acute phase of aSAH may identify patients at risk for unfavorable neurological outcomes and may serve as a marker of early brain injury.
Collapse
|
33
|
Sandrini L, Ieraci A, Amadio P, Zarà M, Barbieri SS. Impact of Acute and Chronic Stress on Thrombosis in Healthy Individuals and Cardiovascular Disease Patients. Int J Mol Sci 2020; 21:ijms21217818. [PMID: 33105629 PMCID: PMC7659944 DOI: 10.3390/ijms21217818] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Psychological stress induces different alterations in the organism in order to maintain homeostasis, including changes in hematopoiesis and hemostasis. In particular, stress-induced hyper activation of the autonomic nervous system and hypothalamic–pituitary–adrenal axis can trigger cellular and molecular alterations in platelets, coagulation factors, endothelial function, redox balance, and sterile inflammatory response. For this reason, mental stress is reported to enhance the risk of cardiovascular disease (CVD). However, contrasting results are often found in the literature considering differences in the response to acute or chronic stress and the health condition of the population analyzed. Since thrombosis is the most common underlying pathology of CVDs, the comprehension of the mechanisms at the basis of the association between stress and this pathology is highly valuable. The aim of this work is to give a comprehensive review of the studies focused on the role of acute and chronic stress in both healthy individuals and CVD patients, focusing on the cellular and molecular mechanisms underlying the relationship between stress and thrombosis.
Collapse
Affiliation(s)
- Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (P.A.); (M.Z.)
- Correspondence: (L.S.); (S.S.B.); Tel.: +39-02-58002021 (L.S. & S.S.B.)
| | - Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Sezione di Fisiologia e Farmacologia, University of Milan, 20133 Milan, Italy;
| | - Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (P.A.); (M.Z.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (P.A.); (M.Z.)
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (P.A.); (M.Z.)
- Correspondence: (L.S.); (S.S.B.); Tel.: +39-02-58002021 (L.S. & S.S.B.)
| |
Collapse
|
34
|
Amadio P, Zarà M, Sandrini L, Ieraci A, Barbieri SS. Depression and Cardiovascular Disease: The Viewpoint of Platelets. Int J Mol Sci 2020; 21:E7560. [PMID: 33066277 PMCID: PMC7589256 DOI: 10.3390/ijms21207560] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Depression is a major cause of morbidity and low quality of life among patients with cardiovascular disease (CVD), and it is now considered as an independent risk factor for major adverse cardiovascular events. Increasing evidence indicates not only that depression worsens the prognosis of cardiac events, but also that a cross-vulnerability between the two conditions occurs. Among the several mechanisms proposed to explain this interplay, platelet activation is the more attractive, seeing platelets as potential mirror of the brain function. In this review, we dissected the mechanisms linking depression and CVD highlighting the critical role of platelet behavior during depression as trigger of cardiovascular complication. In particular, we will discuss the relationship between depression and molecules involved in the CVD (e.g., catecholamines, adipokines, lipids, reactive oxygen species, and chemokines), emphasizing their impact on platelet activation and related mechanisms.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy;
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| |
Collapse
|
35
|
Abstract
The review states that antidepressants (ADs) increase brain-derived neurotrophic factor (BDNF) transmission concomitantly in the brain and the blood: ADs increasing BDNF synthesis in specific areas of the central nervous system (CNS) could presumably affect megakaryocyte's production of platelets. ADs increase BDNF levels in the CNS and improve mood. In the blood, ADs increase BDNF release from platelets. The hypothesis presented here is that the release of BDNF from platelets contributes to the ADs effects on neurogenesis and on tumor growth in the cancer disease. Oncological studies indicate that chemicals ADs exert an aggravating effect on the cancer disease, possibly by promoting proplatelets formation and enhancing BDNF release from platelets in the tumor.
Collapse
Affiliation(s)
- Francis Lavergne
- Physiopathologie des maladies Psychiatriques, Institut de Psychiatrie et Neurosciences de Paris, UMR_S 1266 INSERM, Paris, France
| | - Therese M Jay
- Physiopathologie des maladies Psychiatriques, Institut de Psychiatrie et Neurosciences de Paris, UMR_S 1266 INSERM, Paris, France.,Faculté de Médecine Paris Descartes, Université Paris Descartes, Paris, France
| |
Collapse
|
36
|
Silczuk A, Habrat B. Alcohol-induced thrombocytopenia: Current review. Alcohol 2020; 86:9-16. [PMID: 32330589 DOI: 10.1016/j.alcohol.2020.02.166] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/27/2019] [Accepted: 02/07/2020] [Indexed: 12/11/2022]
Abstract
Thrombocytopenia is a decrease in the platelet count below 150,000 in a microliter of blood, i.e., below the lower limit of the reference range, which is 150,000-400,000/μL. The phenomenon of thrombocytopenia related to heavy drinking began to arouse interest in the 1960s and 1970s. It was initially described in case reports and clinical studies on small groups. In the following years, the phenomenon itself and the significance of alcohol-induced thrombocytopenia was studied. Many methodological difficulties inhibiting objective conclusions from research were encountered. Model pathological mechanisms of alcohol thrombocytopenia and the effects of alcohol on the structure and function of platelets were described. Furthermore, the phenomenon of rapid normalization of the number of platelets in people who stopped drinking was described. Relationships between alcohol use, its intensity and occurrence, and intensity of thrombocytopenia have been demonstrated. Predictive platelet counts for alcohol withdrawal syndrome complications have been proven and calculated. The risk of occurrence of withdrawal seizures or delirium tremens in alcohol withdrawal syndrome increases significantly when the platelet count is less than 119,000/μL. The knowledge of the nature of the phenomenon of alcohol-induced thrombocytopenia in a clinical environment allows decisions that are more rational. The attention of clinicians should be drawn to the importance of results of blood tests routinely collected on admission.
Collapse
|
37
|
Dysregulated megakaryocyte distribution associated with nestin + mesenchymal stem cells in immune thrombocytopenia. Blood Adv 2020; 3:1416-1428. [PMID: 31053569 DOI: 10.1182/bloodadvances.2018026690] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 03/07/2019] [Indexed: 01/28/2023] Open
Abstract
Impaired megakaryocyte (MK) maturation and reduced platelet production are important causes of immune thrombocytopenia (ITP). However, MK distribution and bone marrow (BM) niche alteration in ITP are unclear. To investigate the maturation and distribution of MKs in the BM niche and examine the components of BM niche regulation of MK migration, BM and peripheral blood were obtained from 30 ITP patients and 28 healthy donors. Nestin+ mesenchymal stem cells (MSCs) and CD41+ MKs were sorted by fluorescence-activated cell sorting. The components of the BM niche and related signaling were analyzed via immunofluorescence, flow cytometry, enzyme-linked immunosorbent assay, reverse transcription polymerase chain reaction, and western blot analysis. The number of MKs in the BM vascular niche was reduced in ITP. Moreover, the concentrations of CXCL12 and CXCR4+ MKs in the BM were decreased in ITP. Further investigation demonstrated that nestin+ MSCs and CXCL12 messenger RNA (mRNA) in nestin+ MSCs were both reduced whereas the apoptosis of nestin+ MSCs was significantly increased in ITP. Sympathetic nerves, Schwann cells, the proportion of β3-adrenoreceptor (β3-AR)+ nestin+ MSCs, and β3-AR mRNA in nestin+ MSCs were all markedly reduced in ITP. Moreover, matrix metalloproteinase 9, vascular endothelial growth factor (VEGF), and VEGF receptor 1 were significantly reduced in ITP. Our data show that impaired MK distribution mediated by an abnormal CXCL12/CXCR4 axis is partially involved in reduced platelet production in ITP. Moreover, sympathetic neuropathy and nestin+ MSC apoptosis may have an effect on the alterations of BM CXCL12 in ITP.
Collapse
|
38
|
Chen S, Qi Y, Wang S, Xu Y, Shen M, Hu M, Du C, Chen F, Chen M, Lu Y, Zhang Z, Quan Y, Wang C, Wang F, Wang J. Melatonin enhances thrombopoiesis through ERK1/2 and Akt activation orchestrated by dual adaptor for phosphotyrosine and 3-phosphoinositides. J Pineal Res 2020; 68:e12637. [PMID: 32052470 DOI: 10.1111/jpi.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 11/29/2022]
Abstract
Melatonin (MT), endogenously secreted by the pineal gland, is closely related to multiple biological processes; however, its effect on thrombopoiesis is still not well illustrated. Here, we demonstrate that MT administration can elevate peripheral platelet levels. Analysis of different stages in thrombopoiesis reveals that MT has the capacity to promote the expansion of CD34+ and CD41+ cells, and accelerate proplatelet formation (PPF) and platelet production. Furthermore, in vivo experiments show that MT has a potential therapeutic effect on radiation-induced thrombocytopenia. The underlying mechanism suggests that both extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt signaling are involved in the processes of thrombopoiesis facilitated by MT. Interestingly, in addition to the direct regulation of Akt signaling by its upstream phosphoinositide 3-kinase (PI3K), ERK1/2 signaling is also regulated by PI3K via its effector, dual adaptor for phosphotyrosine and 3-phosphoinositides (DAPP1), in megakaryocytes after MT treatment. Moreover, the expression level of DAPP1 during megakaryocyte differentiation is closely related to the activation of ERK1/2 and Akt at different stages of thrombopoiesis. In conclusion, our data suggest that MT treatment can promote thrombopoiesis, which is modulated by the DAPP1-orchestrated activation of ERK1/2 and Akt signaling.
Collapse
Affiliation(s)
- Shilei Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yan Qi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Song Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yang Xu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Mingqiang Shen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Mengjia Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Fang Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Mo Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yukai Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Zihao Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yong Quan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
39
|
Ho YH, Méndez-Ferrer S. Microenvironmental contributions to hematopoietic stem cell aging. Haematologica 2020; 105:38-46. [PMID: 31806690 PMCID: PMC6939521 DOI: 10.3324/haematol.2018.211334] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell (HSC) aging was originally thought to be essentially an HSC-autonomous process, which is the focus of another review in the same issue of Haematologica However, studies on the microenvironment that maintains and regulates HSC (HSC niche) over the past 20 years have suggested that microenvironmental aging contributes to declined HSC function over time. The HSC niches comprise a complex and dynamic molecular network of interactions across multiple cell types, including endothelial cells, mesenchymal stromal cells, osteoblasts, adipocytes, neuroglial cells and mature hematopoietic cells. Upon aging, functional changes in the HSC niches, such as microenvironmental senescence, imbalanced bone marrow mesenchymal stromal cell differentiation, vascular remodeling, changes in adrenergic signaling and inflammation, coordinately and dynamically influence the fate of HSC and their downstream progeny. The end result is lymphoid deficiency and myeloid skewing. During this process, aged HSC and their derivatives remodel the niche to favor myeloid expansion. Therefore, the crosstalk between HSC and the microenvironment is indispensable for the aging of the hematopoietic system and might represent a therapeutic target in age-related pathological disorders.
Collapse
Affiliation(s)
- Ya-Hsuan Ho
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Simón Méndez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
40
|
Abstract
Secondary hypertension is associated with increased cardiovascular risk and exaggerated target organ damage, not only due to the higher and more sustained blood pressure values often observed in these patients but also because certain forms of hypertension can increase cardiovascular risk and organ damage by the neurohormonal and/or molecular pathways activation they exert. Early identification of secondary forms of hypertension can help mitigate organ damage and prevent cardiovascular complications. Signs and symptoms distinction among types of secondary hypertension is essential in order to prevent complications.
Collapse
Affiliation(s)
- Oscar H Cingolani
- Division of Cardiology, Hypertension Program, Johns Hopkins University Hospital, 601 North Caroline Street, Outpatient Center, 7th Floor, Suite 7263, Baltimore, MD 21287, USA.
| |
Collapse
|
41
|
Ho YH, Del Toro R, Rivera-Torres J, Rak J, Korn C, García-García A, Macías D, González-Gómez C, Del Monte A, Wittner M, Waller AK, Foster HR, López-Otín C, Johnson RS, Nerlov C, Ghevaert C, Vainchenker W, Louache F, Andrés V, Méndez-Ferrer S. Remodeling of Bone Marrow Hematopoietic Stem Cell Niches Promotes Myeloid Cell Expansion during Premature or Physiological Aging. Cell Stem Cell 2019; 25:407-418.e6. [PMID: 31303548 PMCID: PMC6739444 DOI: 10.1016/j.stem.2019.06.007] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 02/21/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem cells (HSCs) residing in the bone marrow (BM) accumulate during aging but are functionally impaired. However, the role of HSC-intrinsic and -extrinsic aging mechanisms remains debated. Megakaryocytes promote quiescence of neighboring HSCs. Nonetheless, whether megakaryocyte-HSC interactions change during pathological/natural aging is unclear. Premature aging in Hutchinson-Gilford progeria syndrome recapitulates physiological aging features, but whether these arise from altered stem or niche cells is unknown. Here, we show that the BM microenvironment promotes myelopoiesis in premature/physiological aging. During physiological aging, HSC-supporting niches decrease near bone but expand further from bone. Increased BM noradrenergic innervation promotes β2-adrenergic-receptor(AR)-interleukin-6-dependent megakaryopoiesis. Reduced β3-AR-Nos1 activity correlates with decreased endosteal niches and megakaryocyte apposition to sinusoids. However, chronic treatment of progeroid mice with β3-AR agonist decreases premature myeloid and HSC expansion and restores the proximal association of HSCs to megakaryocytes. Therefore, normal/premature aging of BM niches promotes myeloid expansion and can be improved by targeting the microenvironment.
Collapse
Affiliation(s)
- Ya-Hsuan Ho
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Raquel Del Toro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBER-CV), Spain
| | - José Rivera-Torres
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBER-CV), Spain
| | - Justyna Rak
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Claudia Korn
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Andrés García-García
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - David Macías
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Cristina González-Gómez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBER-CV), Spain
| | - Alberto Del Monte
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBER-CV), Spain
| | - Monika Wittner
- INSERM (Institut National de la Santé et de la Recherche Médicale), Université Paris-Saclay, UMR1170, Gustave Roussy, 94805 Villejuif, France; Université Paris-Saclay and CNRS GDR 3697 MicroNiT, Villejuif, France
| | - Amie K Waller
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Holly R Foster
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, 33006 Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Randall S Johnson
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Cedric Ghevaert
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - William Vainchenker
- INSERM (Institut National de la Santé et de la Recherche Médicale), Université Paris-Saclay, UMR1170, Gustave Roussy, 94805 Villejuif, France
| | - Fawzia Louache
- INSERM (Institut National de la Santé et de la Recherche Médicale), Université Paris-Saclay, UMR1170, Gustave Roussy, 94805 Villejuif, France; Université Paris-Saclay and CNRS GDR 3697 MicroNiT, Villejuif, France
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares (CIBER-CV), Spain
| | - Simón Méndez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0PT, UK; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK; Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain.
| |
Collapse
|
42
|
Heber S, Fischer B, Sallaberger-Lehner M, Hausharter M, Ocenasek H, Gleiss A, Fischer MJM, Pokan R, Assinger A, Volf I. Effects of high-intensity interval training on platelet function in cardiac rehabilitation: a randomised controlled trial. Heart 2019; 106:69-79. [DOI: 10.1136/heartjnl-2019-315130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/14/2019] [Accepted: 06/18/2019] [Indexed: 12/26/2022] Open
Abstract
ObjectiveTo compare effects of moderate-intensity continuous training (MICT) and high-intensity interval training (HIIT) on platelet function in patients undergoing cardiac rehabilitation, as hyper-reactive platelets are involved in atherogenesis and atherothrombosis.MethodsIn this single-centre parallel group randomised controlled trial, male patients after an acute coronary syndrome under dual antiplatelet therapy performed MICT or HIIT+MICT for 12 weeks. Main outcome was platelet reactivity measured by the half-maximal concentration (EC50) of platelet agonist thrombin receptor-activating peptide-6 (TRAP-6) in terms of P-selectin expression. EC50 was determined at baseline, after 6 and 12 weeks, each time at physical rest and on exertion.Results82 patients were randomised to MICT or HIIT+MICT. Mean (95% CI) baseline EC50values at physical rest were 6.7 µM (6.3 µM to 7.0 µM) TRAP-6. After 6/12 weeks, 36/33 MICT and 34/28 HIIT+MICT patients were examined. HIIT+MICT patients had 0.9 µM (0.4 µM to 1.4 µM)/0.5 µM (−0.1 µM to 1.0 µM) higher EC50values than MICT ones, and the propensity of their platelets to form aggregates with monocytes was significantly lower after 12 weeks. Short-term strenuous physical exertion was generally associated with platelet activation and an EC50reduction of 0.7 µM (0.6 µM to 0.8 µM). HIIT+MICT patients tended to be fitter after 12 weeks. No serious harms were observed.ConclusionsIncluding HIIT in cardiac rehabilitation seems to confer additional benefits compared with MICT alone, which should be confirmed in clinical trials with hard endpoints. Exertion-induced platelet activation and hyper-reactivity occur despite dual antiplatelet therapy.Trial registration numberNCT02930330; Results.
Collapse
|
43
|
Fu W, Meng G, Yang X, Yu L, Jiang H. Bone marrow sympathetic activation regulates post-myocardial infarction megakaryocyte expansion but not platelet production. Biochem Biophys Res Commun 2019; 513:99-104. [DOI: 10.1016/j.bbrc.2019.03.160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 03/24/2019] [Indexed: 10/27/2022]
|
44
|
Silczuk A, Habrat B, Lew-Starowicz M. Thrombocytopenia in Patients Hospitalized for Alcohol Withdrawal Syndrome and Its Associations to Clinical Complications. Alcohol Alcohol 2019; 54:503-509. [PMID: 31403690 DOI: 10.1093/alcalc/agz061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The aim of this study was to assess the predictive value of thrombocytopenia (TP) in alcohol withdrawal syndrome (AWS) as a marker of evolution of non-complicated AWS (nAWS) to severe, complicated AWS (cAWS): delirium tremens (DTs) and withdrawal seizures (wS), and to broaden knowledge about differences between nAWS and cAWS groups in relation to severity of TP. METHODS This study involved 300 people (236 males and 64 females), aged 19-65 years (M = 44.64, SD = 11.32), hospitalized in the detoxification ward with ICD-10 diagnosis of F10.3 (AWS) or F10.4 (DTs), divided into nAWS and cAWS groups, 150 cases each. AWS severity was measured by CIWA-Ar. Available clinical and laboratory data were analyzed. RESULTS TP was found in 139 (46%) of all subjects (nAWS = 32, cAWS = 107). nAWS and cAWS did not differ according to age, gender, length and severity of the last binge. A relationship between the occurrence of TP and cAWS was found (P < 0.001). The lower was the number of PLT, the more AWS incidence was observed. In CIWA-Ar, TP subjects had at least moderate AWS (P < 0.001). nAWS had higher PLT values than cAWS cases (Mrang = 195.96 vs. 105.04, P < 0.001). The predictive value of TP in cAWS was confirmed. CONCLUSIONS The study demonstrates that patients with AWS and TP (in particular <119k/mL) are at higher risk of developing cAWS.
Collapse
Affiliation(s)
- Andrzej Silczuk
- Department of Prevention and Treatment of Addictions, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Bogusław Habrat
- Department of Prevention and Treatment of Addictions, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Michał Lew-Starowicz
- Department of Psychiatry, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
45
|
Gómez-Moreno D, Adrover JM, Hidalgo A. Neutrophils as effectors of vascular inflammation. Eur J Clin Invest 2018; 48 Suppl 2:e12940. [PMID: 29682731 DOI: 10.1111/eci.12940] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Abstract
Vascular inflammation underlies most forms of cardiovascular disease, which remains a prevalent cause of death among the global population. Advances in the biology of neutrophils, as well as insights into their dynamics in tissues, have revealed that these cells are prominent drivers of vascular inflammation though derailed activation within blood vessels. The development of powerful imaging techniques, as well as identification of cells and molecules that regulate their activation within vessels, including platelets and catecholamines, has been instrumental to better understand the mechanisms through which neutrophils protect or damage the organism. Other advances in our understanding of how these leucocytes exert detrimental functions on neighbouring cells, including the formation of DNA-based extracellular traps, constitute milestones in defining neutrophil-driven inflammation. Here, we review emerging mechanisms that regulate intravascular activation and effector functions of neutrophils, and discuss specific pathologies in which these processes are relevant. We argue that identification of pathways and mechanisms specifically engaged within the vasculature may provide effective therapies to treat this prevalent group of pathologies.
Collapse
Affiliation(s)
- Diego Gómez-Moreno
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José María Adrover
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Andrés Hidalgo
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
46
|
Sub-Chronic Stress Exacerbates the Pro-Thrombotic Phenotype in BDNF Val/Met Mice: Gene-Environment Interaction in the Modulation of Arterial Thrombosis. Int J Mol Sci 2018; 19:ijms19103235. [PMID: 30347685 PMCID: PMC6214083 DOI: 10.3390/ijms19103235] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/15/2018] [Accepted: 10/17/2018] [Indexed: 12/21/2022] Open
Abstract
Brain-Derived Neurotrophic Factor (BDNF) Val66Met polymorphism has been associated with increased susceptibility to develop mood disorders and recently it has been also linked with cardiovascular disease (CVD). Interestingly, stressful conditions unveil the anxious/depressive-like behavioral phenotype in heterozygous BDNFVal66Met (BDNFVal/Met) mice, suggesting an important relationship in terms of gene-environment interaction (GxE). However, the interplay between stress and BDNFVal/Met in relation to CVD is completely unknown. Here, we showed that BDNFVal/Met mice display a greater propensity to arterial thrombosis than wild type BDNFVal/Val mice after 7 days of restraint stress (RS). RS markedly increased the number of leukocytes and platelets, and induced hyper-responsive platelets as showed by increased circulating platelet/leukocyte aggregates and enhanced expression of P-selectin and GPIIbIIIa in heterozygous mutant mice. In addition, stressed BDNFVal/Met mice had a greater number of large and reticulated platelets but comparable number and maturation profile of bone marrow megakaryocytes compared to BDNFVal/Val mice. Interestingly, RS led to a significant reduction of BDNF expression accompanied by an increased activity of tissue factor in the aorta of both BDNFVal/Val and BDNFVal/Met mice. In conclusion, we provide evidence that sub-chronic stress unveils prothrombotic phenotype in heterozygous BDNF Val66Met mice affecting both the number and functionality of blood circulating cells, and the expression of key thrombotic molecules in aorta. Human studies will be crucial to understand whether this GxE interaction need to be taken into account in risk stratification of coronary artery disease (CAD) patients.
Collapse
|
47
|
IGF-1 facilitates thrombopoiesis primarily through Akt activation. Blood 2018; 132:210-222. [DOI: 10.1182/blood-2018-01-825927] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022] Open
Abstract
Key Points
IGF-1 has the ability to promote megakaryocyte differentiation, PPF, and platelet release. The effect of IGF-1 on thrombopoiesis is mediated primarily by AKT activation with the assistance of SRC-3.
Collapse
|
48
|
Mirabegron, a β 3-adrenoceptor agonist reduced platelet aggregation through cyclic adenosine monophosphate accumulation. Eur J Pharmacol 2018; 829:79-84. [PMID: 29654782 DOI: 10.1016/j.ejphar.2018.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 11/22/2022]
Abstract
Mirabegron is a β3-adrenoceptor agonist and released on the marked for the treatment of overactive bladder. Because mirabegron is the only β3-adrenoceptor agonist available and substances that increase the levels of cyclic adenosine monophosphate (cAMP) inhibit platelet activity, we tested the hypothesis that mirabegron could have antiplatelet activity. Collagen- and thrombin induced platelet aggregation, thromboxane B2 (TXB2) and cyclic nucleotides quantification and calcium (Ca2+) mobilization were determined in the absence and presence of mirabegron in human washed platelets. Our results revealed that mirabegron (10-300 µM) produced significant inhibitions on platelet aggregation induced by collagen- or thrombin, accompanied by greater intracellular levels of cAMP. The β3-adrenoceptor antagonist L 748,337 (1 µM) and the adenylate cyclase inhibitor, SQ 22,536 (100 µM) reversed the inhibition induced by mirabegron in thrombin-stimulated platelets. The selective antagonists for β1-and β2-adrenoceptors, atenolol and ICI 117,551 (3 µM), respectively did not interfere on the inhibition induced by mirabegron. In Fluo-4 loaded platelets, mirabegron reduced the total and intracellular Ca2+ levels. Pre-incubation with mirabegron almost abolished the levels of TXB2. Mirabegron did not augment the intracellular levels of cyclic guanosine monophosphate. In conclusion, mirabegron inhibited human platelet aggregation through cAMP accumulation, thus suggesting that substances that activate β3-adrenoceptor could be beneficial as adjuvant antiplatelet therapy.
Collapse
|
49
|
Ding S, Wang M, Fang S, Xu H, Fan H, Tian Y, Zhai Y, Lu S, Qi X, Wei F, Sun G, Sun X. D-dencichine Regulates Thrombopoiesis by Promoting Megakaryocyte Adhesion, Migration and Proplatelet Formation. Front Pharmacol 2018; 9:297. [PMID: 29666579 PMCID: PMC5891617 DOI: 10.3389/fphar.2018.00297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Life-threatening chemotherapy-induced thrombocytopenia can increase the risk of bleeding due to a dramatic low platelet count, which may limit or delay treatment schedules in cancer patients. The pressing need for the rapid alleviation of the symptoms of thrombocytopenia has prompted us to search for novel highly effective and safe thrombopoietic agents. Pharmacological investigations have indicated that dencichine can prevent and treat blood loss and increase the number of platelets. On the basis of the neurotoxicity of dencichine, D-dencichine is artificially synthesized in the laboratory. Our initial results showed that D-dencichine had potential to elevate peripheral platelet levels in mice with carboplatin-induced thrombocytopenia. However, the mechanisms of D-dencichine on thrombopoiesis have been poorly understood. In this study, we found that sequential administration of D-dencichine had a distinct ability to elevate numbers of reticulated platelets, and did not alter their clearance. Moreover, we demonstrated that D-dencichine was able to modulate the return of hematopoietic factors to normal levels, including thrombopoietin and IL-6. However, subsequent analysis revealed that D-dencichine treatment had no direct effects on megakaryocytes proliferation, differentiation, and polyploidization. Further in vitro studies, we demonstrated for the first time that D-dencichine significantly stimulated megakaryocyte adhesion, migration, and proplatelet formation in a dose-dependent manner through extracellular regulated protein kinases1/2 (ERK1/2) and v-akt murine thymoma viral oncogene homolog (AKT) signaling pathways. This study sufficiently characterized the role of the effects of D-dencichine treatment on the regulation of thrombopoiesis and provided a promising avenue for CIT treating.
Collapse
Affiliation(s)
- Shilan Ding
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Song Fang
- Kunming Shenghuo Pharmaceutical Group Co., Ltd., Kunming, China
| | - Huibo Xu
- Academy of Chinese Medical Sciences of Jilin Province, Jilin, China
| | - Huiting Fan
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yadong Zhai
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Shan Lu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xin Qi
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fei Wei
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
50
|
He Y, Xu LL, Feng FE, Wang QM, Zhu XL, Wang CC, Zhang JM, Fu HX, Xu LP, Liu KY, Huang XJ, Zhang XH. Mesenchymal stem cell deficiency influences megakaryocytopoiesis through the TNFAIP3/NF-κB/SMAD pathway in patients with immune thrombocytopenia. Br J Haematol 2018; 180:395-411. [PMID: 29327472 DOI: 10.1111/bjh.15034] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/19/2017] [Indexed: 12/27/2022]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease. Mesenchymal stem cells (MSCs) play important roles in the physiology and homeostasis of the haematopoietic system, including supporting megakaryocytic differentiation from CD34+ haematopoietic progenitor cells. Tumour necrosis factor alpha-induced protein 3 (TNFAIP3, also termed A20) plays a key role in terminating NF-κB signalling. Human genetic studies showed that the polymorphisms of the TNFAIP3 gene may contribute to ITP susceptibility. In this study, we showed a significant decrease in TNFAIP3 and increase in NF-κB/SMAD7 in ITP-MSCs. In co-cultures with CD34+ cells, NF-κB was overexpressed in MSCs from healthy controls (HC-MSCs) after transfection with NFKBIA (IκB)-specific short hairpin (sh)RNAs, resulting in MSC deficiency and a reduction in megakaryocytic differentiation and thrombopoiesis. Knockdown of TNFAIP3 expression using TNFAIP3-specific shRNAs in HC-MSCs affected megakaryocytopoiesis. However, IKBKB knockdown corrected megakaryocytopoiesis inhibition in the ITP-MSCs by decreasing NF-κB expression. Amplified TNFAIP3 expression in ITP-MSCs by TNFAIP3 cDNA can facilitate megakaryocyte differentiation. shRNA-mediated knockdown of SMAD7 expression rescued the impaired MSC function in ITP patients. Therefore, we demonstrate that a pathological reduction in TNFAIP3 levels induced NF-κB/SMAD7 pathway activation, causing a deficiency in MSCs in ITP patients. The ability of ITP-MSCs to support megakaryocytic differentiation and thrombopoiesis of CD34+ cells was impaired.
Collapse
Affiliation(s)
- Yun He
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China.,Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Haematology, Peking University, Beijing, China
| | - Lin-Lin Xu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Fei-Er Feng
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Qian-Ming Wang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Xiao-Lu Zhu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Chen-Cong Wang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Jia-Min Zhang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China.,Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Haematology, Peking University, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China.,Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Haematology, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China.,Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Haematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Haematology, Beijing, China.,Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Haematology, Peking University, Beijing, China
| |
Collapse
|