101
|
Aw NH, Canetti E, Suzuki K, Goh J. Monocyte Subsets in Atherosclerosis and Modification with Exercise in Humans. Antioxidants (Basel) 2018; 7:antiox7120196. [PMID: 30572572 PMCID: PMC6315962 DOI: 10.3390/antiox7120196] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/05/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a progressive pathological remodeling of the arteries and one of its hallmarks is the presence of chronic inflammation. Notably, there is an increased proportion and activation state of specific monocyte subsets in systemic blood circulation. Monocyte subsets have distinct contributions to the formation, progression, and destabilization of the atherosclerotic plaque. Strong clinical and epidemiological studies show that regular aerobic exercise mitigates the progression of cardiovascular disease. In fact, aerobic fitness is a powerful predictor of cardiovascular mortality in adults, independent of traditional risk factors such as hypertension and hyperlipidemia. Acute bouts and chronic exercise training modulate monocyte behavior, ranging from their recruitment from the bone marrow or marginal pool, to tissue margination and functional changes in cytokine and chemokine production. Such modulation could reflect a potential mechanism for the cardio-protective effect of exercise on atherosclerosis. This review summarizes the current knowledge of monocyte subsets and highlights what is known about their responses to exercise.
Collapse
Affiliation(s)
- Ning Hong Aw
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore 139651, Singapore.
| | - Elisa Canetti
- Faculty of Health Sciences and Medicine, Bond University, Robina QLD 4226, Australia.
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan.
| | - Jorming Goh
- Ageing Research Institute for Society & Education (ARISE), Nanyang Technological University, Singapore 639798, Singapore.
- Exercise Medicine & Physiology Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore.
| |
Collapse
|
102
|
Ortiz-Rivero S, Baquero C, Hernández-Cano L, Roldán-Etcheverry JJ, Gutiérrez-Herrero S, Fernández-Infante C, Martín-Granado V, Anguita E, de Pereda JM, Porras A, Guerrero C. C3G, through its GEF activity, induces megakaryocytic differentiation and proplatelet formation. Cell Commun Signal 2018; 16:101. [PMID: 30567575 PMCID: PMC6299959 DOI: 10.1186/s12964-018-0311-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Megakaryopoiesis allows platelet formation, which is necessary for coagulation, also playing an important role in different pathologies. However, this process remains to be fully characterized. C3G, an activator of Rap1 GTPases, is involved in platelet activation and regulates several differentiation processes. METHODS We evaluated C3G function in megakaryopoiesis using transgenic mouse models where C3G and C3GΔCat (mutant lacking the GEF domain) transgenes are expressed exclusively in megakaryocytes and platelets. In addition, we used different clones of K562, HEL and DAMI cell lines with overexpression or silencing of C3G or GATA-1. RESULTS We found that C3G participates in the differentiation of immature hematopoietic cells to megakaryocytes. Accordingly, bone marrow cells from transgenic C3G, but not those from transgenic C3GΔCat mice, showed increased expression of the differentiation markers CD41 and CD61, upon thrombopoietin treatment. Furthermore, C3G overexpression increased the number of CD41+ megakaryocytes with high DNA content. These results are supported by data obtained in the different models of megakaryocytic cell lines. In addition, it was uncovered GATA-1 as a positive regulator of C3G expression. Moreover, C3G transgenic megakaryocytes from fresh bone marrow explants showed increased migration from the osteoblastic to the vascular niche and an enhanced ability to form proplatelets. Although the transgenic expression of C3G in platelets did not alter basal platelet counts, it did increase slightly those induced by TPO injection in vivo. Moreover, platelet C3G induced adipogenesis in the bone marrow under pathological conditions. CONCLUSIONS All these data indicate that C3G plays a significant role in different steps of megakaryopoiesis, acting through a mechanism dependent on its GEF activity.
Collapse
Affiliation(s)
- Sara Ortiz-Rivero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Cristina Baquero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Luis Hernández-Cano
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain
| | - Juan José Roldán-Etcheverry
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, IdISSC, Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Gutiérrez-Herrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Cristina Fernández-Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain
| | - Víctor Martín-Granado
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Eduardo Anguita
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, IdISSC, Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - José María de Pereda
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), Universidad de Salamanca-CSIC, Salamanca, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain. .,Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain. .,Centro de Investigación del Cáncer, Campus Unamuno s/n, Salamanca, Spain.
| |
Collapse
|
103
|
Machado-Neto JA, Fenerich BA, Rodrigues Alves APN, Fernandes JC, Scopim-Ribeiro R, Coelho-Silva JL, Traina F. Insulin Substrate Receptor (IRS) proteins in normal and malignant hematopoiesis. Clinics (Sao Paulo) 2018; 73:e566s. [PMID: 30328953 PMCID: PMC6169455 DOI: 10.6061/clinics/2018/e566s] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
The insulin receptor substrate (IRS) proteins are a family of cytoplasmic proteins that integrate and coordinate the transmission of signals from the extracellular to the intracellular environment via transmembrane receptors, thus regulating cell growth, metabolism, survival and proliferation. The PI3K/AKT/mTOR and MAPK signaling pathways are the best-characterized downstream signaling pathways activated by IRS signaling (canonical pathways). However, novel signaling axes involving IRS proteins (noncanonical pathways) have recently been identified in solid tumor and hematologic neoplasm models. Insulin receptor substrate-1 (IRS1) and insulin receptor substrate-2 (IRS2) are the best-characterized IRS proteins in hematologic-related processes. IRS2 binds to important cellular receptors involved in normal hematopoiesis (EPOR, MPL and IGF1R). Moreover, the identification of IRS1/ABL1 and IRS2/JAK2V617F interactions and their functional consequences has opened a new frontier for investigating the roles of the IRS protein family in malignant hematopoiesis. Insulin receptor substrate-4 (IRS4) is absent in normal hematopoietic tissues but may be expressed under abnormal conditions. Moreover, insulin receptor substrate-5 (DOK4) and insulin receptor substrate-6 (DOK5) are linked to lymphocyte regulation. An improved understanding of the signaling pathways mediated by IRS proteins in hematopoiesis-related processes, along with the increased development of agonists and antagonists of these signaling axes, may generate new therapeutic approaches for hematological diseases. The scope of this review is to recapitulate and review the evidence for the functions of IRS proteins in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- João Agostinho Machado-Neto
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
- Departamento de Farmacologia do Instituto de Ciencias Biomedicas da Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bruna Alves Fenerich
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
| | - Ana Paula Nunes Rodrigues Alves
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
| | - Jaqueline Cristina Fernandes
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
| | - Renata Scopim-Ribeiro
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
| | - Juan Luiz Coelho-Silva
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
| | - Fabiola Traina
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| |
Collapse
|
104
|
Ribatti D, Tamma R. Hematopoietic growth factors and tumor angiogenesis. Cancer Lett 2018; 440-441:47-53. [PMID: 30312730 DOI: 10.1016/j.canlet.2018.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/26/2018] [Accepted: 10/08/2018] [Indexed: 01/13/2023]
Abstract
Angiogenesis is regulated by numerous "classic" factors such as vascular endothelial growth factor (VEGF) and many other endogenous "non-classic"peptides, including erythropoietin (Epo), and granulocyte-/granulocyte macrophage colony stimulating factor (G-/GM-CSF). The latter play an important regulatory role in angiogenesis, especially under pathological conditions and constitute a crosslink between angiogenesis and hematopoiesis. This article reviews studies on the ability of hematopoietic cytokines to affect several endothelial cell functions in tumor angiogenesis. These findings in all these studies support the hypothesis formulated at the beginning of this century that a common ancestral cell, the hemangioblast, gives rise to cells of both the endothelial and the hematopoietic lineages.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
105
|
Fisher MH, Di Paola J. Genomics and transcriptomics of megakaryocytes and platelets: Implications for health and disease. Res Pract Thromb Haemost 2018; 2:630-639. [PMID: 30349880 PMCID: PMC6178711 DOI: 10.1002/rth2.12129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/03/2018] [Indexed: 01/07/2023] Open
Abstract
The field of megakaryocyte and platelet biology has been transformed with the implementation of high throughput sequencing. The use of modern sequencing technologies has led to the discovery of causative mutations in congenital platelet disorders and has been a useful tool in uncovering many other mechanisms of altered platelet formation and function. Although the understanding of the presence of RNA in platelets is relatively novel, mRNA and miRNA expression profiles are being shown to play an increasingly important role in megakaryopoiesis and platelet function in normal physiology as well as in disease states. Understanding the genetic perturbations underlying platelet dysfunction provides insight into normal megakaryopoiesis and thrombopoiesis, as well as guiding the development of novel therapeutics.
Collapse
Affiliation(s)
- Marlie H. Fisher
- Department of PediatricsUniversity of Colorado School of MedicineAuroraColorado
- Medical Scientist Training ProgramUniversity of Colorado School of MedicineAuroraColorado
| | - Jorge Di Paola
- Department of PediatricsUniversity of Colorado School of MedicineAuroraColorado
- Medical Scientist Training ProgramUniversity of Colorado School of MedicineAuroraColorado
| |
Collapse
|
106
|
Gupta T, Puskarich MA, DeVos E, Javed A, Smotherman C, Sterling SA, Wang HE, Moore FA, Jones AE, Guirgis FW. Sequential Organ Failure Assessment Component Score Prediction of In-hospital Mortality From Sepsis. J Intensive Care Med 2018; 35:810-817. [PMID: 30165769 DOI: 10.1177/0885066618795400] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Early organ dysfunction in sepsis confers a high risk of in-hospital mortality, but the relative contribution of specific types of organ failure to overall mortality is unclear. The objective of this study was to assess the predictive ability of individual types of organ failure to in-hospital mortality or prolonged intensive care. METHODS Retrospective cohort study of adult emergency department patients with sepsis from October 1, 2013, to November 10, 2015. Multivariable regression was used to assess the odds ratios of individual organ failure types for the outcomes of in-hospital death (primary) and in-hospital death or ICU stay ≥ 3 days (secondary). RESULTS Of 2796 patients, 283 (10%) experienced in-hospital mortality, and 748 (27%) experienced in-hospital mortality or an ICU stay ≥ 3 days. The following components of Sequential Organ Failure Assessment (SOFA) score were most predictive of in-hospital mortality (descending order): coagulation (odds ratio [OR]: 1.60, 95% confidence interval [CI]: 1.32-1.93), hepatic (1.58, 95% CI: 1.32-1.90), respiratory (OR: 1.33, 95% CI: 1.21-1.47), neurologic (OR: 1.20, 95% CI: 1.07-1.35), renal (OR: 1.14, 95% CI: 1.02-1.27), and cardiovascular (OR: 1.13, 95% CI: 1.01-1.25). For mortality or ICU stay ≥3 days, the most predictive SOFA components were respiratory (OR: 1.97, 95% CI: 1.79-2.16), neurologic (OR: 1.72, 95% CI: 1.54-1.92), cardiovascular (OR: 1.38, 95% CI: 1.23-1.54), coagulation (OR: 1.31, 95% CI: 1.10-1.55), and renal (OR: 1.19, 95% CI: 1.08-1.30) while hepatic SOFA (OR: 1.16, 95% CI: 0.98-1.37) did not reach statistical significance (P = .092). CONCLUSION In this retrospective study, SOFA score components demonstrated varying predictive abilities for mortality in sepsis. Elevated coagulation or hepatic SOFA scores were most predictive of in-hospital death, while an elevated respiratory SOFA was most predictive of death or ICU stay >3 days.
Collapse
Affiliation(s)
- Tushar Gupta
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| | - Michael A Puskarich
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Elizabeth DeVos
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| | - Adnan Javed
- Department of Emergency Medicine, University of Massachusetts, Boston, MA, USA
| | - Carmen Smotherman
- Center for Health Equity and Quality Research, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Sarah A Sterling
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Henry E Wang
- Department of Emergency Medicine, University of Texas, Austin, TX, USA
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alan E Jones
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| |
Collapse
|
107
|
Islam J, Zaman K, Chakrabarti S, Chattopadhyay P. Exploration of toxicological impacts following acute and sub-chronic exposure to ethyl anthranilate-loaded mosquito repellent patch. Regul Toxicol Pharmacol 2018; 97:209-224. [PMID: 29969653 DOI: 10.1016/j.yrtph.2018.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 02/05/2018] [Accepted: 06/29/2018] [Indexed: 12/12/2022]
Abstract
In the recent years, growing concern about the potential toxicity of synthetic repellents has led to the development of environmentally safe non-toxic insect control methods. Present investigation explores the toxicological impacts of ethyl anthranilate-loaded mosquito repellent patch (EAMRP) on respiratory system following acute and sub-chronic inhalation exposure in Wistar rats. Lungs parameters such as enhanced pause, tidal volume, respiration rate, inspiration time, and expiration time were determined using whole body plethysmograph. X-ray, scanning electron microscopy and histology were utilized to study the morphology and microscopical architecture of lungs. Hematological and serum biochemical markers were estimated. Cytokines such as IL-1β, IL-2, and IL-12 were also estimated in bronchoalveolar lavage fluid using ELISA kits. Finally, acute oral and dermal toxicity studies were carried out to study the accidental or intentional poisoning due to the ingestion and skin contact of EAMRP, respectively. The findings demonstrate that inhalation exposure to EAMRP did not pose any significant dose related toxicity in above mentioned experiments. Further, no appreciable toxicity was observed in both acute oral and dermal exposure. Thus, these results revealed the non toxic nature of EAMRP in preclinical studies. Hence, EAMRP can be used successfully as an alternative to existing synthetic repellents without any potential health hazards.
Collapse
Affiliation(s)
- Johirul Islam
- Defence Research Laboratory, Tezpur, Assam, 784001, India; Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Kamaruz Zaman
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | | | | |
Collapse
|
108
|
Takeda T, Morita H, Saito H, Matsumoto K, Matsuda A. Recent advances in understanding the roles of blood platelets in the pathogenesis of allergic inflammation and bronchial asthma. Allergol Int 2018; 67:326-333. [PMID: 29242144 DOI: 10.1016/j.alit.2017.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/12/2017] [Accepted: 11/19/2017] [Indexed: 12/28/2022] Open
Abstract
Platelets play an essential role in hemostasis to minimize blood loss due to traumatic injury. In addition, they contain various immune-associated molecules and contribute to immunological barrier formation at sites of vascular injury, thereby protecting against invading pathogens. Platelets are also crucially involved in development of allergic diseases, including bronchial asthma. Platelets in asthmatics are more activated than those in healthy individuals. By using a murine asthma model, platelets were shown to be actively involved in progression of the disease, including in airway eosinophilia and airway remodeling. In the asthmatic airway, pathological microvascular angiogenesis, a component of airway remodeling, is commonly observed, and the degree of abnormality is significantly associated with disease severity. Therefore, in order to repair the newly formed and structurally fragile blood vessels under inflammatory conditions, platelets may be continuously activated in asthmatics. Importantly, platelets constitutively express IL-33 protein, an alarmin cytokine that is essential for development of bronchial asthma. Meanwhile, the concept of development of allergic diseases has recently changed dramatically, and allergy researchers now share a belief in the centrality of epithelial barrier functions. In particular, IL-33 released from epithelial barrier tissue at sites of eczema can activate the antigen-non-specific innate immune system as an alarmin that is believed to be necessary for subsequent antigen-specific acquired immunological responses. From this perspective, we propose in this review a possible mechanism for how activated platelets act as an alarmin in development of bronchial asthma.
Collapse
Affiliation(s)
- Tomohiro Takeda
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Health Sciences, Kansai University of Health Sciences, Osaka, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akio Matsuda
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
109
|
Colic L, Woelfer M, Colic M, Leutritz AL, Liebe T, Fensky L, Sen ZD, Li M, Hoffmann J, Kretzschmar MA, Isermann B, Walter M. Delayed increase of thrombocyte levels after a single sub-anesthetic dose of ketamine - A randomized trial. Eur Neuropsychopharmacol 2018; 28:701-709. [PMID: 29699723 DOI: 10.1016/j.euroneuro.2018.03.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 03/07/2018] [Accepted: 03/22/2018] [Indexed: 12/25/2022]
Abstract
Recently, ketamine has been investigated as a potential antidepressant option for treatment resistant depression. Unlike traditional drugs, it yields immediate effects, most likely via increased glutamatergic transmission and synaptic plasticity. However, ketamine administration in humans is systemic and its long-term impact on blood parameters has not yet been described in clinical studies. Here we investigated potential sustained effects of ketamine administration (0.5 mg/kg ketamine racemate) on hematological and biochemical values in plasma and serum in a randomized double-blinded study. 80 healthy young participants were included and whole blood samples were collected 5 days before, and 14 days after the infusion. To assess the group effect, repeated measure analyses of co-variance (rmANCOVA) were conducted for the following blood parameters: levels of sodium, potassium, calcium, hemoglobin and number of erythrocytes, lymphocytes, and thrombocytes. RmANCOVA revealed a significant time by treatment effect on thrombocyte levels (F1, 74 = 13.54, p < 0.001, eta = 0.155), driven by an increase in the ketamine group (paired t-test, t = -3.51, df = 38, p = 0.001). Specificity of thrombocyte effect was confirmed by logistic regression, and in addition, no other coagulation parameters showed significant interaction. Moreover, the relative increase in the ketamine group was stable across sexes and not predicted by age, BMI, smoking, alcohol or drug use, and contraception. Our results describe aftereffects of sub-anesthetic ketamine administration on blood coagulation parameters, which should be considered especially when targeting psychiatric populations with relevant clinical comorbidities.
Collapse
Affiliation(s)
- Lejla Colic
- Clinical Affective Neuroimaging Laboratory, Germany; Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, Germany; Otto-von-Guericke-University Magdeburg, Magdeburg, Germany; Brain Connectivity Laboratory, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, USA
| | - Merima Colic
- Department of Laboratory Diagnostics, Children's Hospital Zagreb, Zagreb, Croatia
| | - Anna Linda Leutritz
- Clinical Affective Neuroimaging Laboratory, Germany; Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Thomas Liebe
- Clinical Affective Neuroimaging Laboratory, Germany
| | - Luisa Fensky
- Clinical Affective Neuroimaging Laboratory, Germany; Translational Psychiatry, Department of Psychiatry, University of Tübingen, Tübingen, Germany
| | - Zumrut Duygu Sen
- Clinical Affective Neuroimaging Laboratory, Germany; Translational Psychiatry, Department of Psychiatry, University of Tübingen, Tübingen, Germany
| | - Meng Li
- Clinical Affective Neuroimaging Laboratory, Germany; Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Juliane Hoffmann
- Institute for Clinical chemistry and Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Moritz A Kretzschmar
- Department of Anaesthesiology and Intensive Care Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Berend Isermann
- Institute for Clinical chemistry and Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, Germany; Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany; Translational Psychiatry, Department of Psychiatry, University of Tübingen, Tübingen, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany; Max Planck Institute for Biological Cybernetics, Tübingen, Germany.
| |
Collapse
|
110
|
Scherlinger M, Guillotin V, Truchetet ME, Contin-Bordes C, Sisirak V, Duffau P, Lazaro E, Richez C, Blanco P. Systemic lupus erythematosus and systemic sclerosis: All roads lead to platelets. Autoimmun Rev 2018; 17:625-635. [PMID: 29635077 DOI: 10.1016/j.autrev.2018.01.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 01/18/2018] [Indexed: 01/13/2023]
Abstract
Systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) are two phenotypically distincts inflammatory systemic diseases. However, SLE and SSc share pathogenic features such as interferon signature, loss of tolerance against self-nuclear antigens and increased tissue damage such as fibrosis. Recently, platelets have emerged as a major actor in immunity including auto-immune diseases. Both SLE and SSc are characterized by strong platelet system activation, which is likely to be both the witness and culprit in their pathogenesis. Platelet activation pathways are multiple and sometimes redundant. They include immune complexes, Toll-like receptors activation, antiphospholipid antibodies and ischemia-reperfusion associated with Raynaud phenomenon. Once activated, platelet promote immune dysregulation by priming interferon production by immune cells, providing CD40L supporting B lymphocyte functions and providing a source of autoantigens. Platelets are actively implicated in SLE and SSc end-organ damage such as cardiovascular and renal disease and in the promotion of tissue fibrosis. Finally, after understanding the main pathogenic implications of platelet activation in both diseases, we discuss potential therapeutics targeting platelets.
Collapse
Affiliation(s)
- Marc Scherlinger
- Service de Rhumatologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Vivien Guillotin
- Service de médecine interne, FHU ACRONIM, Hôpital Saint André, Centre Hospitalier Universitaire, 1 rue Jean Burguet, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Marie-Elise Truchetet
- Service de Rhumatologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Cécile Contin-Bordes
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Vanja Sisirak
- Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Pierre Duffau
- Service de médecine interne, FHU ACRONIM, Hôpital Saint André, Centre Hospitalier Universitaire, 1 rue Jean Burguet, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Estibaliz Lazaro
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Christophe Richez
- Service de Rhumatologie, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Patrick Blanco
- Laboratoire d'Immunologie et Immunogénétique, FHU ACRONIM, Hôpital Pellegrin, Centre Hospitalier Universitaire, Place Amélie Raba Léon, 33076 Bordeaux, France; Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS-UMR 5164, ImmunoConcept, Université de Bordeaux, 146 rue Léo Saignat, 33076 Bordeaux, France.
| |
Collapse
|
111
|
|
112
|
Abstract
Thrombopoietin (THPO) has been well characterized as a key regulator of platelet production. THPO also plays an important role in the maintenance and regulation of hematopoietic stem cells (HSCs). In this issue of EMBO Molecular Medicine, Pecci et al (2018) describe a newly identified homozygous mutation in THPO causing congenital amegakaryocytic thrombocytopenia, a disease characterized by a significant impairment in platelet production with rapid onset of aplastic anemia within a few years. The paper nicely investigates the underlying pathogenic mechanisms of this disease. Importantly, this study, in tandem with other recent ones, shows that this rare genetic form of aplastic anemia is treatable with THPO receptor agonists, emphasizing the paramount role of genetic testing in cases of aplastic anemia and other bone marrow failure disorders. This report also refines our understanding of the role of THPO in human HSC function and illustrates the important biological insight that can be gained through studies of such rare genetic disorders.
Collapse
Affiliation(s)
- Ah Ram Kim
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
113
|
Joo KH, Song JS, Kim S, Lee HS, Jeon M, Kim SO, Lee JH. Cytokine Expression of Stem Cells Originating from the Apical Complex and Coronal Pulp of Immature Teeth. J Endod 2017; 44:87-92.e1. [PMID: 29079048 DOI: 10.1016/j.joen.2017.08.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/05/2017] [Accepted: 08/15/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The aim of this study was to measure and compare the expression levels of cytokines from developing apical complex cells (DACCs) and dental pulp stem cells (DPSCs) of the immature tooth. METHODS DPSC-conditioned medium (CM) and DACCs-CM were obtained from human young teeth, and 174 cytokines secreted from each CM were identified and compared. A cytokine membrane array and enzyme-linked immunosorbent assay were used to measure and compare the expression levels of the cytokines. Immunocytochemistry targeting insulin-like growth factor-1 and neurotrophin-3 was additionally performed. RESULTS There were statistically significant differences in the expression levels of 25 cytokines: 22 and 3 were expressed more strongly in DPSCs-CM and DACCs-CM, respectively. Odontoblast differentiation-related cytokines were more strongly expressed in DPSCs-CM, while cell-proliferation-related cytokines were more strongly expressed in DACCs-CM. Proinflammatory and anti-inflammatory cytokines were predominantly expressed in DPSCs-CM and DACCs-CM, respectively. CONCLUSIONS DPSCs may exert a stronger paracrine effect than DACCs on regeneration of the dentin-pulp complex, in terms of odontoblast differentiation.
Collapse
Affiliation(s)
- Ki Hoon Joo
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Je Seon Song
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea; Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Seunghye Kim
- Department of Pediatric Dentistry, Institute of Oral Health Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyo-Seol Lee
- Department of Pediatric Dentistry, Dental School, Kyung Hee University, Seoul, Republic of Korea
| | - Mijeong Jeon
- Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Seong-Oh Kim
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea; Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea
| | - Jae-Ho Lee
- Department of Pediatric Dentistry, College of Dentistry, Yonsei University, Seoul, Republic of Korea; Oral Science Research Center, College of Dentistry, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
114
|
Rjiba-Touati K, Amara I, Bousabbeh M, Salem IB, Azzebi A, Guedri Y, Achour A, Bacha H, Abid S. Recombinant human erythropoietin prevents etoposide- and methotrexate-induced toxicity in kidney and liver tissues via the regulation of oxidative damage and genotoxicity in Wistar rats. Hum Exp Toxicol 2017; 37:848-858. [PMID: 29069929 DOI: 10.1177/0960327117733553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Etoposide (ETO) and methotrexate (MTX) are two effective chemotherapeutic drugs. However, the clinical use of these drugs is limited by its toxicity in normal tissues, especially in kidney and in liver tissues. Recombinant human erythropoietin (rhEPO), erythropoietin hormone, has also been shown to exert tissue protective effects. The purpose of this study was to explore the protective effect of rhEPO against oxidative stress and genotoxicity induced by ETO and MTX in vivo. Adult male Wistar rats were divided into 10 groups (6 animals each): control group, rhEPO alone group, ETO alone group, MTX alone group and rhEPO + ETO/MTX groups. In rhEPO + ETO/MTX groups, three doses of pretreatment with rhEPO were performed: 1000, 3000 and 6000 IU/kg. Our results showed that rhEPO pretreatment protects liver and kidney tissues against oxidative stress induced by the anticancer drugs. The glycoprotein decreased malondialdehyde (MDA) levels, reduced catalase activity and ameliorated glutathione depletion. Furthermore, we showed that rhEPO administration prevented drug-induced DNA damage accessed by comet test. Altogether, our results suggested a protective role of rhEPO, especially at 3000 IU/kg, against ETO- and MTX-induced oxidative stress and genotoxicity in vivo.
Collapse
Affiliation(s)
- K Rjiba-Touati
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Amara
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - M Bousabbeh
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - I Ben Salem
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - A Azzebi
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - Y Guedri
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - A Achour
- 2 Department of Nephrology, Dialysis and Transplant, University Hospital of Sahloul, Sousse, Tunisia
| | - H Bacha
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| | - S Abid
- 1 Laboratory of Research on Biologically Compatible Compounds, Faculty of Dentistry, Monastir University, Monastir, Tunisia
| |
Collapse
|
115
|
Myles N, Myles H, Clark SR, Bird R, Siskind D. Use of granulocyte-colony stimulating factor to prevent recurrent clozapine-induced neutropenia on drug rechallenge: A systematic review of the literature and clinical recommendations. Aust N Z J Psychiatry 2017; 51:980-989. [PMID: 28747065 DOI: 10.1177/0004867417720516] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Clozapine is the most effective medication for treatment-refractory schizophrenia; however, its use is contraindicated in people who have had previous clozapine-induced neutropenia. Co-prescription of granulocyte-colony stimulating factor may prevent recurrent neutropenia and allow continuation or rechallenge of clozapine. OBJECTIVE AND METHODS Systematic review of literature reporting the use of granulocyte-colony stimulating factor to allow rechallenge or continuation of clozapine in people with previous episodes of clozapine-induced neutropenia. The efficacy of granulocyte-colony stimulating factor and predictors of successful rechallenge will be determined to elucidate whether evidence-based recommendations can be made regarding the use of granulocyte-colony stimulating factor in this context. RESULTS A total of 17 articles were identified that reported on clozapine rechallenge with granulocyte-colony stimulating factor support. In all, 76% of cases were able to continue clozapine at median follow-up of 12 months. There were no clear clinical or laboratory predictors of successful rechallenge; however, initial neutropenia was more severe in successful cases compared to unsuccessful cases. Cases co-prescribed lithium had lower success rates of rechallenge (60%) compared to those who were not prescribed lithium (81%). The most commonly reported rechallenge strategy was use of filgrastim 150-480 µg between daily to three times a week. There were no medication-specific side effects of granulocyte-colony stimulating factor reported apart from euphoria in one case. Three cases who failed granulocyte-colony stimulating factor had bacterial infection at time of recurrent neutropenia. No deaths were reported. CONCLUSION Preliminary data suggest granulocyte-colony stimulating factor is safe and effective in facilitating rechallenge with clozapine. Clinical recommendations for use are discussed.
Collapse
Affiliation(s)
- Nicholas Myles
- 1 Haematology Directorate, SA Pathology, Adelaide, SA, Australia.,2 School of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Hannah Myles
- 3 Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia.,4 Country Health SA, Glenside, SA, Australia
| | - Scott R Clark
- 3 Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Robert Bird
- 5 Division of Cancer Services, Princess Alexandra Hospital, Wooloongabba, QLD, Australia.,6 School of Medicine, Griffith University, Nathan, QLD, Australia
| | - Dan Siskind
- 2 School of Medicine, University of Queensland, St Lucia, QLD, Australia.,7 Metro South Addiction and Mental Health Service, Brisbane, QLD, Australia
| |
Collapse
|
116
|
Dodillet H, Kreuzer K, Monsef I, Skoetz N. Thrombopoietin mimetics for patients with myelodysplastic syndromes. Cochrane Database Syst Rev 2017; 9:CD009883. [PMID: 28962071 PMCID: PMC6483680 DOI: 10.1002/14651858.cd009883.pub2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Myelodysplastic syndrome (MDS) is one of the most frequent haematologic malignancies of the elderly population and characterised by progenitor cell dysplasia with ineffective haematopoiesis and a high rate of transformation to acute myeloid leukaemia (AML). Thrombocytopenia represents a common problem for patients with MDS. ranging from mild to serious bleeding events and death. To manage thrombocytopenia, the current standard treatment includes platelet transfusion, unfortunately leading to a range of side effects. Thrombopoietin (TPO) mimetics represent an alternative treatment option for MDS patients with thrombocytopenia. However, it remains unclear, whether TPO mimetics influence the increase of blast cells and therefore to premature progression to AML. OBJECTIVES To evaluate the efficacy and safety of thrombopoietin (TPO) mimetics for patients with MDS. SEARCH METHODS We searched for randomised controlled trials in the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE (January 2000 to August 2017), trials registries (ISRCTN, EU clinical trials register and clinicaltrials.gov) and conference proceedings. We did not apply any language restrictions. Two review authors independently screened search results, disagreements were solved by discussion. SELECTION CRITERIA We included randomised controlled trials comparing TPO mimetics with placebo, no further treatment or another TPO mimetic in patients with MDS of all risk groups, without gender, age or ethnicity restrictions. Additional chemotherapeutic treatment had to be equal in both arms. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed the quality of trials, disagreements were resolved by discussion. Risk ratio (RR) was used to analyse mortality during study, transformation to AML, incidence of bleeding events, transfusion requirement, all adverse events, adverse events >= grade 3, serious adverse events and platelet response. Overall survival (OS) and progression-free survival (PFS) have been extracted as hazard ratios, but could not be pooled as results were reported in heterogenous ways. Health-related quality of life and duration of thrombocytopenia would have been analysed as standardised mean differences, but no trial reported these outcomes. MAIN RESULTS We did not identify any trial comparing one TPO mimetic versus another. We analysed six eligible trials involving 746 adult patients. All trials were reported as randomised and double-blind trials including male and female patients. Two trials compared TPO mimetics (romiplostim or eltrombopag) with placebo, one trial evaluated eltrombopag in addition to the hypomethylating agent azacitidine, two trials analysed romiplostim additionally to a hypomethylating agent (azacitidine or decitabine) and one trial evaluated romiplostim in addition to the immunomodulatory drug lenalidomide. There are more data on romiplostim (four included, completed, full-text trials) than on eltrombopag (two trials included: one full-text publication, one abstract publication). Due to small sample sizes and imbalances in baseline characteristics in three trials and premature termination of two studies, we judged the potential risk of bias of all included trials as high.Due to heterogenous reporting, we were not able to pool data for OS. Instead of that, we analysed mortality during study. There is little or no evidence for a difference in mortality during study for thrombopoietin mimetics compared to placebo (RR 0.97, 95% confidence interval (CI) 0.73 to 1.27, N = 6 trials, 746 patients, low-quality evidence). It is unclear whether the use of TPO mimetics induces an acceleration of transformation to AML (RR 1.02, 95% CI 0.59 to 1.77, N = 5 trials, 372 patients, very low-quality evidence).Thrombopoietin mimetics probably improve the incidence of all bleeding events (RR 0.92, 95% CI 0.86 to 0.99, N = 5 trials, 390 patients, moderate-quality evidence). This means that in the study population, 713 out of 1000 in the placebo arm will have a bleeding event, compared to 656 of 1000 (95% CI 613 to 699) in the TPO mimetics arm. There is little or no evidence for a difference that TPO mimetics significantly diminish the rate of transfusion requirement (RR 0.83, 95% CI 0.66 to 1.05, N = 4 trials, 358 patients, low-quality evidence). No studies were found that looked at quality of life or duration of thrombocytopenia.There is no evidence that patients given TPO mimetics suffer more all adverse events (RR 1.01, 95% CI 0.96 to 1.07, N = 5 trials, 390 patients, moderate-quality evidence). There is uncertainty whether the number of serious adverse events decrease under therapy with TPO mimetics (RR 0.89, 95% CI 0.54 to 1.46, N = 4 trials, 356 patients, very low-quality evidence).We identified one ongoing study and one study marked as completed (March 2015), but without publication of results for MDS patients (only results reported for AML and MDS patients together). Both studies evaluate MDS patients receiving eltrombopag in comparison to placebo. AUTHORS' CONCLUSIONS No trial evaluated one TPO mimetic versus another.Six trials including adult patients analysed one TPO mimetic versus placebo, sometimes combined with standard therapy in both arms. Given the uncertainty of the quality of evidence, meta-analyses show that there is little or no evidence for a difference in mortality during study and premature progress to AML. However, these assumptions have to be further explored. Treatment with TPO mimetics resulted in a lower number of MDS patients suffering from bleeding events.There is no evidence for a difference between study groups regarding transfusion requirement. Enlarged sample sizes and a longer follow-up of future trials should improve the estimate of safety and efficacy of TPO mimetics, moreover health-related quality of life should be evaluated. As two ongoing studies currently investigate eltrombopag (one already completed, but without published results), we are awaiting results for this drug.
Collapse
Affiliation(s)
- Helga Dodillet
- University Hospital of CologneDepartment I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Karl‐Anton Kreuzer
- University Hospital of CologneDepartment I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Ina Monsef
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Nicole Skoetz
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | | |
Collapse
|
117
|
The Effect of Granulocyte Colony-Stimulating Factor on the Progression of Atherosclerosis in Animal Models: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6705363. [PMID: 29138752 PMCID: PMC5613364 DOI: 10.1155/2017/6705363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 07/12/2017] [Indexed: 11/17/2022]
Abstract
Background Atherosclerosis is a common inflammatory disease. Stem cell and endothelial progenitor cell treatments can improve cardiac function after myocardial infarction. Granulocyte colony-stimulating factor (G-CSF) is a mobilisation agent, mobilising stem cells from the bone marrow to circulation in the blood. G-CSF may constitute a treatment of atherosclerosis. We have conducted meta-analysis to evaluate the current evidence for the effect of G-CSF on the progression of atherosclerosis in animal models and to provide reference for preclinical experiments and future human clinical trials of atherosclerosis treatment. Methods We searched several databases and conducted a meta-analysis across seven articles using a random-effect model. All statistical analyses were performed using Review Manager Version 5.2 and Stata 12.0. Results We found that G-CSF therapy was associated with reduced atherosclerotic lesion area (weighted mean difference (WMD): 7.29%; 95% confidence interval (CI): 2.06-12.52%; P = 0.006). No significant differences in total serum cholesterol (P = 0.54) and triglyceride levels (P = 0.95) were noted in G-CSF treatment groups compared with controls. Multivariable metaregression analysis revealed that the animal type (rabbit, P = 0.022) and frequency of G-CSF administration (>20, P = 0.007) impacted the atherosclerotic lesion area changes. Conclusion The meta-analysis suggested that G-CSF treatment might inhibit the progression of atherosclerosis in animal models.
Collapse
|
118
|
Misra H, Berryman J, Jubin R, Abuchowski A. A Phase I study to determine safety, pharmacokinetics, and pharmacodynamics of ANF-RHO™, a novel PEGylated granulocyte colony-stimulating factor, in healthy volunteers. Invest New Drugs 2017; 36:75-84. [PMID: 28752433 DOI: 10.1007/s10637-017-0490-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/05/2017] [Indexed: 11/25/2022]
Abstract
Patients receiving pegfilgrastim (Neulasta®) for the treatment of neutropenia can experience bone pain following the injections required to achieve effective neutrophil levels. The safety, pharmacokinetic (PK), and pharmacodynamic (PD) profiles of ANF-RHO™, a novel pegylated granulocyte colony stimulating factor, were assessed in a randomized, controlled, double-blind Phase 1 clinical study in healthy volunteers. Subjects received a single subcutaneous dose of ANF-RHO over a range of 6 doses (5-50 μg/kg), placebo (saline), or the recommended clinical dose of pegfilgrastim administered at the labeled fixed 6 mg dosage (equivalent to 80-100 μg/kg). The primary outcome measure was safety and tolerability. Secondary outcomes included PK and PD effects on absolute neutrophil count (ANC) and number of CD34+ progenitor cells. Severity of bone pain was also assessed. In healthy volunteers, ANF-RHO was administered at ascending doses up to 50 μg/kg without significant adverse effects; appeared to be better (5 to 30 μg/kg) or equally well (50 μg/kg) tolerated, and had lower mean bone pain scores as compared to pegfilgrastim. ANF-RHO achieved CD34+ and ANC numbers at significantly lower doses, and had a significantly longer circulating half-life than pegfilgrastim. These results suggest that ANF-RHO can be provided less frequently, at a lower dose, and with fewer side effects. ANF-RHO had unique, prolonged PK/PD attributes as compared to marketed pegfilgrastim, suggesting that it may provide an improved clinical benefit in further clinical studies in patients with chemotherapy-induced or chronic idiopathic neutropenia.
Collapse
Affiliation(s)
- Hemant Misra
- Prolong Pharmaceuticals, 300 Corporate Court, Suite B, South Plainfield, NJ, 07080, USA.
| | - John Berryman
- Prolong Pharmaceuticals, 300 Corporate Court, Suite B, South Plainfield, NJ, 07080, USA
| | - Ronald Jubin
- Prolong Pharmaceuticals, 300 Corporate Court, Suite B, South Plainfield, NJ, 07080, USA
| | - Abraham Abuchowski
- Prolong Pharmaceuticals, 300 Corporate Court, Suite B, South Plainfield, NJ, 07080, USA
| |
Collapse
|
119
|
Petersen R, Lambourne JJ, Javierre BM, Grassi L, Kreuzhuber R, Ruklisa D, Rosa IM, Tomé AR, Elding H, van Geffen JP, Jiang T, Farrow S, Cairns J, Al-Subaie AM, Ashford S, Attwood A, Batista J, Bouman H, Burden F, Choudry FA, Clarke L, Flicek P, Garner SF, Haimel M, Kempster C, Ladopoulos V, Lenaerts AS, Materek PM, McKinney H, Meacham S, Mead D, Nagy M, Penkett CJ, Rendon A, Seyres D, Sun B, Tuna S, van der Weide ME, Wingett SW, Martens JH, Stegle O, Richardson S, Vallier L, Roberts DJ, Freson K, Wernisch L, Stunnenberg HG, Danesh J, Fraser P, Soranzo N, Butterworth AS, Heemskerk JW, Turro E, Spivakov M, Ouwehand WH, Astle WJ, Downes K, Kostadima M, Frontini M. Platelet function is modified by common sequence variation in megakaryocyte super enhancers. Nat Commun 2017; 8:16058. [PMID: 28703137 PMCID: PMC5511350 DOI: 10.1038/ncomms16058] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/19/2017] [Indexed: 12/26/2022] Open
Abstract
Linking non-coding genetic variants associated with the risk of diseases or disease-relevant traits to target genes is a crucial step to realize GWAS potential in the introduction of precision medicine. Here we set out to determine the mechanisms underpinning variant association with platelet quantitative traits using cell type-matched epigenomic data and promoter long-range interactions. We identify potential regulatory functions for 423 of 565 (75%) non-coding variants associated with platelet traits and we demonstrate, through ex vivo and proof of principle genome editing validation, that variants in super enhancers play an important role in controlling archetypical platelet functions. Numerous genetic variants, including those located in the non-coding regions of the genome, are known to be associated with blood cells traits. Here, Frontini and colleagues investigate their potential regulatory functions using epigenomic data and promoter long-range interactions.
Collapse
Affiliation(s)
- Romina Petersen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - John J Lambourne
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Biola M Javierre
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Roman Kreuzhuber
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Dace Ruklisa
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Medical Research Council Biostatistics Unit, University of Cambridge, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Isabel M Rosa
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Ana R Tomé
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Heather Elding
- Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,Strangeways Research Laboratory, The National Institute for Health Research (NIHR) Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge CB1 8RN, UK
| | - Johanna P van Geffen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Tao Jiang
- Strangeways Research Laboratory, MRC/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Jonathan Cairns
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Abeer M Al-Subaie
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Dammam, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Sofie Ashford
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Antony Attwood
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Joana Batista
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Heleen Bouman
- Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Frances Burden
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Fizzah A Choudry
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Laura Clarke
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Stephen F Garner
- National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Matthias Haimel
- NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.,Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Carly Kempster
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Vasileios Ladopoulos
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - An-Sofie Lenaerts
- NIHR Cambridge Biomedical Research Centre hIPSC Core Facility, Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK.,Wellcome Trust and MRC Cambridge Stem Cell Institute, Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK
| | - Paulina M Materek
- NIHR Cambridge Biomedical Research Centre hIPSC Core Facility, Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK.,Wellcome Trust and MRC Cambridge Stem Cell Institute, Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Stuart Meacham
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Daniel Mead
- Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Christopher J Penkett
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Augusto Rendon
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Genomics England Limited, Queen Mary University of London, Dawson Hall, London EC1M 6BQ, UK
| | - Denis Seyres
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Benjamin Sun
- Strangeways Research Laboratory, MRC/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Salih Tuna
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Marie-Elise van der Weide
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Steven W Wingett
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Joost H Martens
- Faculty of Science, Department of Molecular Biology, Radboud University, 6525GA Nijmegen, The Netherlands
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Sylvia Richardson
- Medical Research Council Biostatistics Unit, University of Cambridge, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Ludovic Vallier
- Wellcome Trust and MRC Cambridge Stem Cell Institute, Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SZ, UK.,The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - David J Roberts
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headington, Oxford OX9 3DU, UK.,Department of Haematology, Churchill Hospital, Headington, Oxford OX3 7LE, UK.,NHSBT, John Radcliffe Hospital, Headington, Oxford OX3 9BQ, UK
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven 3000, Belgium
| | - Lorenz Wernisch
- Medical Research Council Biostatistics Unit, University of Cambridge, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Hendrik G Stunnenberg
- Faculty of Science, Department of Molecular Biology, Radboud University, 6525GA Nijmegen, The Netherlands
| | - John Danesh
- Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,Strangeways Research Laboratory, The National Institute for Health Research (NIHR) Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge CB1 8RN, UK.,Strangeways Research Laboratory, MRC/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.,Department of Biological Science, Florida State University, Tallahassee, Florida 32303, USA
| | - Nicole Soranzo
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,Strangeways Research Laboratory, The National Institute for Health Research (NIHR) Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge CB1 8RN, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Adam S Butterworth
- Strangeways Research Laboratory, The National Institute for Health Research (NIHR) Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge CB1 8RN, UK.,Strangeways Research Laboratory, MRC/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Johan W Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,NIHR BioResource-Rare Diseases, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK.,Medical Research Council Biostatistics Unit, University of Cambridge, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK
| | - Mikhail Spivakov
- Nuclear Dynamics Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK.,Strangeways Research Laboratory, The National Institute for Health Research (NIHR) Blood and Transplant Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge CB1 8RN, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - William J Astle
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,Medical Research Council Biostatistics Unit, University of Cambridge, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, UK.,Strangeways Research Laboratory, MRC/British Heart Foundation (BHF) Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Myrto Kostadima
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge CB2 0PT, UK.,BHF Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| |
Collapse
|
120
|
Yu C, Jiang Z, Hou A, Mu Y, Liu W, Tan S. Shen-Cao granules formulated based on traditional Chinese medicine alleviates bone marrow suppression caused by platinum-based anticancer reagents. Medicine (Baltimore) 2017; 96:e6818. [PMID: 28489759 PMCID: PMC5428593 DOI: 10.1097/md.0000000000006818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The aim of this study was to evaluate effects of Shen-Cao granules for the prevention of thrombocytopenia caused by anticancer chemotherapy. METHODS In this prospective study, a total of 200 patients with various malignant tumors were enrolled and evenly divided into a Shen-Cao granule treatment (n = 100) and a control group (n = 100). After 2 cycles chemotherapy with any combination of platinum-based drugs (cisplatin, carboplatin, and nedaplatin), the blood platelet (PLT) counts, levels of the PLT production regulator thrombopoietin (TPO), PLT aggregation rates, and the PLT activation marker CD62P expressions were monitored for 2 weeks. RESULTS During 2 weeks of post-chemotherapy, the mean values of the minimum PLT count were 49.65 ± 7.35 × 10/L in the treatment group and 31.56 ± 9.32 × 10/L in the control group. The PLT count in the treatment group reached the lowest value 1.8 days later and recovered to a concentration ≥100 × 10/L 3 days earlier than in the control group. The concentrations of the TPO were 71.43 ± 1.74 and 87.24 ± 0.92 ng/mL in the treatment group and 65.75 ± 1.39 and 67.75 ± 0.67 ng/mL in the control group at 7 and 14 days post-chemotherapy, respectively. The maximum PLT aggregation rate declined after chemotherapy in the treatment group from 58.14 ± 11.46% to 52.89 ± 10.52%, while it increased in the control group from 56.94 ± 10.55% to 61.75 ± 12.26%. Coordinately, the expression of CD62P in the treatment group decreased from 6.17 ± 0.59% to 4.89 ± 0.72%, while it increased from 6.09 ± 0.75% to 7.75 ± 0.67% in the control group. CONCLUSION Our study demonstrated that Shen-Cao granule treatment alleviated thrombocytopenia after chemotherapy, and reduced tumor-induced PLT activation and aggregation.
Collapse
Affiliation(s)
| | - Zhonghua Jiang
- Department of Image, Yantai Hospital of Traditional Chinese Medicine, Yantai, China
| | | | | | | | | |
Collapse
|
121
|
Regulation of Inflammation- and Infection-Driven Hematopoiesis. Trends Immunol 2017; 38:345-357. [DOI: 10.1016/j.it.2017.01.004] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/21/2022]
|
122
|
Adebayo AH, Yakubu OF, Adegbite OS, Okubena O. Haematopoietic induction and hepatic protective roles of Hepacare® in CCl4-induced hepatic damaged rats. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s00580-017-2428-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
123
|
Quantification and three-dimensional microanatomical organization of the bone marrow. Blood Adv 2017; 1:407-416. [PMID: 29296956 DOI: 10.1182/bloodadvances.2016003194] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/05/2017] [Indexed: 12/21/2022] Open
Abstract
Bone marrow (BM) constitutes one of the largest organs in mice and humans, continuously generating, in a highly regulated manner, red blood cells, platelets, and white blood cells that together form the majority of cells of the body. In this review, we provide a quantitative overview of BM cellular composition, we summarize emerging knowledge on its structural organization and cellular niches, and we argue for the need of multidimensional approaches such as recently developed imaging techniques to uncover the complex spatial logic that underlies BM function in health and disease.
Collapse
|
124
|
Brain-derived Neurotrophic Factor Is Associated With Disease Severity and Clinical Outcome in Ugandan Children Admitted to Hospital With Severe Malaria. Pediatr Infect Dis J 2017; 36:146-150. [PMID: 27798544 DOI: 10.1097/inf.0000000000001382] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Malaria remains a leading cause of childhood death and neurologic disability in sub-Saharan Africa. Here, we test the hypothesis that malaria-induced alterations to circulating brain-derived neurotrophic factor (BDNF) are associated with poor clinical outcomes in children with severe malaria. METHODS We quantified BDNF (by enzyme-linked immunosorbent assay) in plasma samples collected [at presentation (day 1), day 3 and day 14], during a prospective study of Ugandan children admitted to hospital with severe malaria (n = 179). RESULTS BDNF concentration at presentation (day 1) was lower in children with cerebral malaria (P < 0.01), coma (P < 0.01), Lambaréné Organ Dysfunction Score >1 (P < 0.05) and respiratory distress (P < 0.01). Higher BDNF concentration at presentation was associated with shorter time to coma recovery [hazard ratio = 1.655 (1.194-2.293); P = 0.002] and a reduced odds ratio of disability [0.50 (0.27-0.94); P = 0.047] and death [0.45 (0.22-0.92); P = 0.035]. BDNF concentration was lower on day 1 and increased in children surviving severe malaria (day 14; P < 0.0001). CONCLUSIONS Our findings provide the new evidence linking circulating BDNF with disease severity, coma recovery and clinical outcome in children with severe malaria.
Collapse
|
125
|
Abstract
BACKGROUND Fontan survivors have depressed cardiac index that worsens over time. Serum biomarker measurement is minimally invasive, rapid, widely available, and may be useful for serial monitoring. The purpose of this study was to identify biomarkers that correlate with lower cardiac index in Fontan patients. Methods and results This study was a multi-centre case series assessing the correlations between biomarkers and cardiac magnetic resonance-derived cardiac index in Fontan patients ⩾6 years of age with biochemical and haematopoietic biomarkers obtained ±12 months from cardiac magnetic resonance. Medical history and biomarker values were obtained by chart review. Spearman's Rank correlation assessed associations between biomarker z-scores and cardiac index. Biomarkers with significant correlations had receiver operating characteristic curves and area under the curve estimated. In total, 97 cardiac magnetic resonances in 87 patients met inclusion criteria: median age at cardiac magnetic resonance was 15 (6-33) years. Significant correlations were found between cardiac index and total alkaline phosphatase (-0.26, p=0.04), estimated creatinine clearance (0.26, p=0.02), and mean corpuscular volume (-0.32, p<0.01). Area under the curve for the three individual biomarkers was 0.63-0.69. Area under the curve for the three-biomarker panel was 0.75. Comparison of cardiac index above and below the receiver operating characteristic curve-identified cut-off points revealed significant differences for each biomarker (p<0.01) and for the composite panel [median cardiac index for higher-risk group=2.17 L/minute/m2 versus lower-risk group=2.96 L/minute/m2, (p<0.01)]. CONCLUSIONS Higher total alkaline phosphatase and mean corpuscular volume as well as lower estimated creatinine clearance identify Fontan patients with lower cardiac index. Using biomarkers to monitor haemodynamics and organ-specific effects warrants prospective investigation.
Collapse
|
126
|
|
127
|
Hammadah M, Al Mheid I, Wilmot K, Ramadan R, Abdelhadi N, Alkhoder A, Obideen M, Pimple PM, Levantsevych O, Kelli HM, Shah A, Sun YV, Pearce B, Kutner M, Long Q, Ward L, Ko YA, Hosny Mohammed K, Lin J, Zhao J, Bremner JD, Kim J, Waller EK, Raggi P, Sheps D, Quyyumi AA, Vaccarino V. Telomere Shortening, Regenerative Capacity, and Cardiovascular Outcomes. Circ Res 2016; 120:1130-1138. [PMID: 27956416 PMCID: PMC5376244 DOI: 10.1161/circresaha.116.309421] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/22/2016] [Accepted: 12/12/2016] [Indexed: 01/04/2023]
Abstract
RATIONALE Leukocyte telomere length (LTL) is a biological marker of aging, and shorter LTL is associated with adverse cardiovascular outcomes. Reduced regenerative capacity has been proposed as a mechanism. Bone marrow-derived circulating progenitor cells are involved in tissue repair and regeneration. OBJECTIVE Main objective of this study was to examine the relationship between LTL and progenitor cells and their impact on adverse cardiovascular outcomes. METHODS AND RESULTS We measured LTL by quantitative polymerase chain reaction in 566 outpatients (age: 63±9 years; 76% men) with coronary artery disease. Circulating progenitor cells were enumerated by flow cytometry. After adjustment for age, sex, race, body mass index, smoking status, and previous myocardial infarction, a shorter LTL was associated with a lower CD34+ cell count: for each 10% shorter LTL, CD34+ levels were 5.2% lower (P<0.001). After adjustment for the aforementioned factors, both short LTL (<Q1) and low CD34+ levels (<Q1) predicted adverse cardiovascular outcomes (death, myocardial infarction, coronary revascularization, or cerebrovascular events) independently of each other, with a hazard ratio of 1.8 and 95% confidence interval of 1.1 to 2.0, and a hazard ratio of 2.1 and 95% confidence interval of 1.3 to 3.0, respectively, comparing Q1 to Q2-4. Patients who had both short LTL (<Q1) and low CD34+ cell count (<Q1) had the greatest risk of adverse outcomes (hazard ratio =3.5; 95% confidence interval, 1.7-7.1). CONCLUSIONS Although shorter LTL is associated with decreased regenerative capacity, both LTL and circulating progenitor cell levels are independent and additive predictors of adverse cardiovascular outcomes in coronary artery disease patients. Our results suggest that both biological aging and reduced regenerative capacity contribute to cardiovascular events, independent of conventional risk factors.
Collapse
Affiliation(s)
- Muhammad Hammadah
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Ibhar Al Mheid
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Kobina Wilmot
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Ronnie Ramadan
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Naser Abdelhadi
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Ayman Alkhoder
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Malik Obideen
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Pratik M Pimple
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Oleksiy Levantsevych
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Heval M Kelli
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Amit Shah
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Yan V Sun
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Brad Pearce
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Michael Kutner
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Qi Long
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Laura Ward
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Yi-An Ko
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Kareem Hosny Mohammed
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Jue Lin
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Jinying Zhao
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - J Douglas Bremner
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Jinhee Kim
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Edmund K Waller
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Paolo Raggi
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - David Sheps
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Arshed A Quyyumi
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.)
| | - Viola Vaccarino
- From the Division of Cardiology, Department of Medicine (M.H., I.A.M., K.W., R.R., N.A., A.A., M.O., H.M.K., A.S., K.H.M., A.A.Q., V.V.) and Department of Psychiatry and Behavioral Sciences (J.D.B.), Emory University School of Medicine, Atlanta, GA; Department of Epidemiology, Rollins School of Public Health (P.M.P., O.L., A.S., Y.V.S., B.P., V.V.), Department of Biostatistics and Bioinformatics, Rollins School of Public Health (Y.V.S., M.K., Q.L., L.W., Y.-A.K.), and Department of Hematology and Oncology, Winship Cancer Institute (J.K., E.K.W.), Emory University, Atlanta, GA; Department of Biochemistry and Biophysics, University of California, San Francisco (J.L.); Department of Epidemiology, Tulane University School of Public Health, New Orleans, LA (J.Z.); Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada (P.R.); and Department of Epidemiology, University of Florida, Gainesville (D.S.).
| |
Collapse
|
128
|
Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment. Front Immunol 2016; 7:502. [PMID: 27895645 PMCID: PMC5107568 DOI: 10.3389/fimmu.2016.00502] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022] Open
Abstract
All hematopoietic and immune cells are continuously generated by hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) through highly organized process of stepwise lineage commitment. In the steady state, HSCs are mostly quiescent, while HPCs are actively proliferating and contributing to daily hematopoiesis. In response to hematopoietic challenges, e.g., life-threatening blood loss, infection, and inflammation, HSCs can be activated to proliferate and engage in blood formation. The HSC activation induced by hematopoietic demand is mediated by direct or indirect sensing mechanisms involving pattern recognition receptors or cytokine/chemokine receptors. In contrast to the hematopoietic challenges with obvious clinical symptoms, how the aging process, which involves low-grade chronic inflammation, impacts hematopoiesis remains undefined. Herein, we summarize recent findings pertaining to functional alternations of hematopoiesis, HSCs, and the bone marrow (BM) microenvironment during the processes of aging and inflammation and highlight some common cellular and molecular changes during the processes that influence hematopoiesis and its cells of origin, HSCs and HPCs, as well as the BM microenvironment. We also discuss how age-dependent alterations of the immune system lead to subclinical inflammatory states and how inflammatory signaling might be involved in hematopoietic aging. Our aim is to present evidence supporting the concept of “Inflamm-Aging,” or inflammation-associated aging of hematopoiesis.
Collapse
Affiliation(s)
- Larisa V Kovtonyuk
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Kristin Fritsch
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Xiaomin Feng
- International Research Center for Medical Sciences , Kumamoto , Japan
| | - Markus G Manz
- Division of Hematology, University Hospital Zurich, University of Zurich , Zurich , Switzerland
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences , Kumamoto , Japan
| |
Collapse
|
129
|
Abstract
PURPOSE OF REVIEW During severe systemic infection, steady-state hematopoiesis is switched to demand-adapted myelopoiesis, leading to increased myeloid progenitor proliferation and, depending on the context and type of pathogen, enhanced granulocytic or monocytic differentiation, respectively. We will review the recent advances in understanding direct and indirect mechanisms by which different pathogen signals are detected and subsequently translated into demand-adapted myelopoiesis. RECENT FINDINGS Enhanced myeloid progenitor proliferation and neutrophil differentiation following infection with prototypic Gram-negative bacterium Escherichia coli is mediated by granulocyte colony-stimulating factor, and reactive oxygen species released from endothelial cells and mature myeloid cells, respectively. Furthermore, hematopoietic stem and progenitor cells directly sense pathogen signals via Toll-like receptors and contribute to emergency granulopoiesis via release and subsequent autocrine and paracrine action of myelopoietic cytokines including IL-6. Moreover, emergency monocytopoiesis upon viral infection depends on T cell-derived IFNγ and release of IL-6 from bone marrow stromal cells. SUMMARY A complex picture is evolving in which various hematopoietic and nonhematopoietic cell types interact with the hematopoietic system in an intricate manner to shape an appropriate hematopoietic response to specific infectious stimuli.
Collapse
|
130
|
Rahmig S, Kronstein-Wiedemann R, Fohgrub J, Kronstein N, Nevmerzhitskaya A, Bornhäuser M, Gassmann M, Platz A, Ordemann R, Tonn T, Waskow C. Improved Human Erythropoiesis and Platelet Formation in Humanized NSGW41 Mice. Stem Cell Reports 2016; 7:591-601. [PMID: 27618723 PMCID: PMC5063583 DOI: 10.1016/j.stemcr.2016.08.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 11/20/2022] Open
Abstract
Human erythro-megakaryopoiesis does not occur in humanized mouse models, preventing the in vivo analysis of human hematopoietic stem cell (HSC) differentiation into these lineages in a surrogate host. Here we show that stably engrafted KIT-deficient NOD/SCID Il2rg−/−KitW41/W41 (NSGW41) mice support much improved human erythropoiesis and platelet formation compared with irradiated NSG recipients. Considerable numbers of human erythroblasts and mature thrombocytes are present in the bone marrow and blood, respectively. Morphology, composition, and enucleation capacity of de novo generated human erythroblasts in NSGW41 mice are comparable with those in human bone marrow. Overexpression of human erythropoietin showed no further improvement in human erythrocyte output, but depletion of macrophages led to the appearance of human erythrocytes in the blood. Human erythropoiesis up to normoblasts and platelet formation is fully supported in NSGW41 mice, allowing the analysis of human HSC differentiation into these lineages, the exploration of certain pathophysiologies, and the evaluation of gene therapeutic approaches. High engraftment of human erythro-megakaryocytic cells in NSGW41 mice Complete maturation of human thrombocytes in vivo Robust human erythropoiesis up to final nucleated RBC progenitors in vivo
Collapse
Affiliation(s)
- Susann Rahmig
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Romy Kronstein-Wiedemann
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany; Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, 01307 Dresden, Germany
| | - Juliane Fohgrub
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Nicole Kronstein
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany; Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, 01307 Dresden, Germany
| | - Aleksandra Nevmerzhitskaya
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Martin Bornhäuser
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, 01307 Dresden, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Alexander Platz
- DKMS Cord Blood Bank, Enderstrasse 94, 01277 Dresden, Germany
| | - Rainer Ordemann
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, 01307 Dresden, Germany
| | - Torsten Tonn
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North-East, 01307 Dresden, Germany; Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, 01307 Dresden, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
131
|
Abstract
The protein kinase C (PKC) family, discovered in the late 1970s, is composed of at least 10 serine/threonine kinases, divided into three groups based on their molecular architecture and cofactor requirements. PKC enzymes have been conserved throughout evolution and are expressed in virtually all cell types; they represent critical signal transducers regulating cell activation, differentiation, proliferation, death, and effector functions. PKC family members play important roles in a diverse array of hematopoietic and immune responses. This review covers the discovery and history of this enzyme family, discusses the roles of PKC enzymes in the development and effector functions of major hematopoietic and immune cell types, and points out gaps in our knowledge, which should ignite interest and further exploration, ultimately leading to better understanding of this enzyme family and, above all, its role in the many facets of the immune system.
Collapse
Affiliation(s)
- Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037; ,
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037; ,
| |
Collapse
|
132
|
Hadley KB, Ryan AS, Forsyth S, Gautier S, Salem N. The Essentiality of Arachidonic Acid in Infant Development. Nutrients 2016; 8:216. [PMID: 27077882 PMCID: PMC4848685 DOI: 10.3390/nu8040216] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 01/16/2023] Open
Abstract
Arachidonic acid (ARA, 20:4n-6) is an n-6 polyunsaturated 20-carbon fatty acid formed by the biosynthesis from linoleic acid (LA, 18:2n-6). This review considers the essential role that ARA plays in infant development. ARA is always present in human milk at a relatively fixed level and is accumulated in tissues throughout the body where it serves several important functions. Without the provision of preformed ARA in human milk or infant formula the growing infant cannot maintain ARA levels from synthetic pathways alone that are sufficient to meet metabolic demand. During late infancy and early childhood the amount of dietary ARA provided by solid foods is low. ARA serves as a precursor to leukotrienes, prostaglandins, and thromboxanes, collectively known as eicosanoids which are important for immunity and immune response. There is strong evidence based on animal and human studies that ARA is critical for infant growth, brain development, and health. These studies also demonstrate the importance of balancing the amounts of ARA and DHA as too much DHA may suppress the benefits provided by ARA. Both ARA and DHA have been added to infant formulas and follow-on formulas for more than two decades. The amounts and ratios of ARA and DHA needed in infant formula are discussed based on an in depth review of the available scientific evidence.
Collapse
Affiliation(s)
- Kevin B Hadley
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Alan S Ryan
- Clinical Research Consulting, 9809 Halston Manor, Boynton Beach, FL 33473, USA.
| | - Stewart Forsyth
- School of Medicine, Dentistry & Nursing, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| | - Sheila Gautier
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| | - Norman Salem
- DSM Nutritional Products, 6480 Dobbin Road, Columbia, MD 21045, USA.
| |
Collapse
|
133
|
Serra-Millàs M. Are the changes in the peripheral brain-derived neurotrophic factor levels due to platelet activation? World J Psychiatry 2016; 6:84-101. [PMID: 27014600 PMCID: PMC4804271 DOI: 10.5498/wjp.v6.i1.84] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/08/2015] [Accepted: 01/22/2016] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in central nervous system development, neurogenesis and neuronal plasticity. BDNF is also expressed in several non-neuronal tissues, and it could play an important role in other processes, such as cancer, angiogenesis, etc. Platelets are the major source of peripheral BDNF. However, platelets also contain high amounts of serotonin; they express specific surface receptors during activation, and a multitude of pro-inflammatory and immunomodulatory bioactive compounds are secreted from the granules. Until recently, there was insufficient knowledge regarding the relationship between BDNF and platelets. Recent studies showed that BDNF is present in two distinct pools in platelets, in α-granules and in the cytoplasm, and only the BDNF in the granules is secreted following stimulation, representing 30% of the total BDNF in platelets. BDNF has an important role in the pathophysiology of depression. Low levels of serum BDNF have been described in patients with major depressive disorder, and BDNF levels increased with chronic antidepressant treatment. Interestingly, there is an association between depression and platelet function. This review analyzed studies that evaluated the relationship between BDNF and platelet activation and the effect of treatments on both parameters. Only a few studies consider this possible confounding factor, and it could be very important in diseases such as depression, which show changes in both parameters.
Collapse
|
134
|
Billio A, Morello E, Mian M, Antoniazzi F, Moschetti I, Cinquini M. Granulopoiesis-stimulating factors for preventing infections after autologous peripheral stem cell transplantation for lymphoma and multiple myeloma in adults. Hippokratia 2016. [DOI: 10.1002/14651858.cd010659.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Atto Billio
- Central Hospital S, Maurizio; Department of Haematology and Bone Marrow Transplantation; Bolzano Italy 39100
| | - Enrico Morello
- Spedali Civili di Brescia; Haematology Department; Brescia Italy 25100
| | - Michael Mian
- Central Hospital S, Maurizio; Department of Haematology and Bone Marrow Transplantation; Bolzano Italy 39100
| | | | - Ivan Moschetti
- Mario Negri Institute for Pharmacological Research; Italian Cochrane Centre; Via La Masa, 19 Milan Italy 20156
| | - Michela Cinquini
- Mario Negri; Centro Cochrane Italiano; via Giuseppe La Masa 19 MILANO Italy 20156
| |
Collapse
|
135
|
Rivera FJ, Kazanis I, Ghevaert C, Aigner L. Beyond Clotting: A Role of Platelets in CNS Repair? Front Cell Neurosci 2016; 9:511. [PMID: 26834562 PMCID: PMC4718976 DOI: 10.3389/fncel.2015.00511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/21/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University SalzburgSalzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
| | - Ilias Kazanis
- Department of Clinical Neuroscience, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of CambridgeCambridge, UK; Department of Biology, University of PatrasPatras, Greece
| | - Cedric Ghevaert
- Department of Haematology, University of CambridgeCambridge, UK; National Health Service Blood and Transplant, Cambridge Biomedical CampusCambridge, UK
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University SalzburgSalzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University SalzburgSalzburg, Austria
| |
Collapse
|
136
|
Zhou C, Huang Y, Wang D, An C, Zhou F, Li Y, Chen G, Wu C, He J, Wu G, Song X, Gao J, Liu W, Li B, Shi J, Huang C, Yu J, Feng J, Yue H, Shi M, Xia J. A Randomized Multicenter Phase III Study of Single Administration of Mecapegfilgrastim (HHPG-19K), a Pegfilgrastim Biosimilar, for Prophylaxis of Chemotherapy-Induced Neutropenia in Patients With Advanced Non-Small-Cell Lung Cancer (NSCLC). Clin Lung Cancer 2015; 17:119-27. [PMID: 26781346 DOI: 10.1016/j.cllc.2015.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Mecapegfilgrastim (code name HHPG-19K) is a biosimilar to pegylated recombinant human granulocyte-colony stimulating factor (PEG-rhG-CSF). The efficacy and safety of mecapegfilgrastim, using a regimen of once-per-cycle injection of 100-μg/kg or a fixed 6-mg dose, were evaluated for the prophylactic therapy for neutropenia in patients with advanced non-small-cell lung cancer (NSCLC) who were treated with myelosuppressive chemotherapy. MATERIALS AND METHODS Patients were randomized (1:1:1) blindly to 3 treatment arms to receive a single injection of mecapegfilgrastim 100 μg/kg, a 6-mg fixed dose of mecapegfilgrastim, or saline (control) in cycle 1. In cycles 2 to 4 following unblinding at the end of cycle 1, patients in the control arm received daily injections of short-acting rhG-CSF at a dose of 5 μg/kg, whereas patients in the 2 mecapegfilgrastim arms continued the same treatment as in cycle 1. All patients received 4 chemotherapy cycles of docetaxel combined with cisplatin or carboplatin every 21 days. The primary endpoint was the incidence of grade ≥ 3 neutropenia in cycle 1. RESULTS A single dose of 100 μg/kg or a fixed 6-mg dose of mecapegfilgrastim per cycle effectively reduced chemotherapy-induced neutropenia and was comparable to daily rhG-CSF with regard to all efficacy endpoints, including incidence of grade ≥ 3 neutropenia, incidence of febrile neutropenia, duration of grade ≥ 3 neutropenia, and time to neutrophil recovery. No difference in efficacy parameters was observed between the 2-dose regimens of mecapegfilgrastim across all cycles. Mecapegfilgrastim was well-tolerated and was as safe as daily rhG-CSF. CONCLUSION Once-per-cycle injection of mecapegfilgrastim is as effective and safe as daily rhG-CSF for prophylaxis of chemotherapy-induced neutropenia in patients with NSCLC. Mecapegfilgrastim (fixed 6-mg dose) is recommended in clinical practice for its convenient dose management.
Collapse
Affiliation(s)
- Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Yunchao Huang
- Department of Thoracic Surgery, Cancer Hospital of Yunnan Provience, Kunming, China
| | - Donglin Wang
- Department of Oncology, Chongqing Cancer Hospital, Chongqing, China
| | - Changshan An
- Department of Respiration Medicine, Yanbian University Hospital, Yanbian, China
| | - Fuxiang Zhou
- Department of Radiotherapy and Chemotherapy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yali Li
- Department of Respiration Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Gongyan Chen
- Department of Respiration Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Changping Wu
- Department of Oncology, The First People's Hospital of Changzhou, Changzhou, China
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Gang Wu
- Department of Oncology, Wuhan Union Hospital, Wuhan, China
| | - Xia Song
- Department of Respiration Medicine, Cancer Hospital of Shanxi Provence, Taiyuan, China
| | - Jianfei Gao
- Department of Oncology, Wuhan General Hospital of Guangzhou Military, Wuhan, China
| | - Wei Liu
- Department of Oncology, Tumor Hospital of Hebei Provence, Shijiazhuang, China
| | - Baolan Li
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jianhua Shi
- Department of Oncology, Linyi Cancer Hospital, Linyi, China
| | - Cheng Huang
- Department of Oncology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Jingrui Yu
- Department of Oncology, The Second People's Hospital of Sichuan, Chengdu, China
| | - Jueping Feng
- Department of Oncology, Wuhan Puai Hospital, Wuhan, China
| | - Hongmei Yue
- Department of Respiration Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Meiqi Shi
- Department of Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Jielai Xia
- Department of Statistics, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
137
|
Buchbinder D, Hsieh L, Mahajerin A, Puthenveetil G, Soni A, Nugent D. Successful treatment of secondary graft failure following unrelated cord blood transplant with hematopoietic growth factors in a pediatric patient with Fanconi anemia. Pediatr Transplant 2015; 19:E181-4. [PMID: 26228438 DOI: 10.1111/petr.12564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2015] [Indexed: 01/02/2023]
Abstract
Graft failure following allogeneic HCT in Fanconi anemia is associated with significant mortality. Retransplantation may be considered; however, the limited toxicity profile of HGFs also makes them an option for the treatment of graft failure. We describe a five-yr-old female diagnosed with Fanconi anemia and marrow failure treated with HCT. The course was complicated by secondary graft failure treated successfully with HGFs including G-CSF, EPO, and romiplostim. The outcome could be related to the intervention, but could also be the natural course of recovery, including recovering from a recent CMV infection treated with ganciclovir. We found the use of HGFs to be an effective and safe alternative to the potential complications as well as morbidity and mortality associated with the use of retransplantation.
Collapse
Affiliation(s)
- David Buchbinder
- Division of Hematology, CHOC Children's Hospital, Orange, CA, USA
| | - Loan Hsieh
- Division of Hematology, CHOC Children's Hospital, Orange, CA, USA
| | - Arash Mahajerin
- Division of Hematology, CHOC Children's Hospital, Orange, CA, USA
| | | | - Amit Soni
- Division of Hematology, CHOC Children's Hospital, Orange, CA, USA
| | - Diane Nugent
- Division of Hematology, CHOC Children's Hospital, Orange, CA, USA
| |
Collapse
|
138
|
Calip GS, Malmgren JA, Lee WJ, Schwartz SM, Kaplan HG. Myelodysplastic syndrome and acute myeloid leukemia following adjuvant chemotherapy with and without granulocyte colony-stimulating factors for breast cancer. Breast Cancer Res Treat 2015; 154:133-43. [PMID: 26450505 PMCID: PMC4718738 DOI: 10.1007/s10549-015-3590-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/03/2015] [Indexed: 10/23/2022]
Abstract
Risk of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) post-breast cancer treatment with adjuvant chemotherapy and granulocyte colony-stimulating factors (G-CSF) is not fully characterized. Our objective was to estimate MDS/AML risk associated with specific breast cancer treatments. We conducted a retrospective cohort study of women aged ≥66 years with stage I-III breast cancer between 2001 and 2009 using the Surveillance, Epidemiology, and End Results-Medicare database. Women were classified as receiving treatment with radiation, chemotherapy, and/or G-CSF. We used multivariable Cox proportional hazards models to estimate adjusted hazard ratios (HR) and 95 % confidence intervals (CI) for MDS/AML risk. Among 56,251 breast cancer cases, 1.2 % developed MDS/AML during median follow-up of 3.2 years. 47.1 % of women received radiation and 14.3 % received chemotherapy. Compared to breast cancer cases treated with surgery alone, those treated with chemotherapy (HR = 1.38, 95 %-CI 0.98-1.93) and chemotherapy/radiation (HR = 1.77, 95 %-CI 1.25-2.51) had increased risk of MDS/AML, but not radiation alone (HR = 1.08, 95 % CI 0.86-1.36). Among chemotherapy regimens and G-CSF, MDS/AML risk was differentially associated with anthracycline/cyclophosphamide-containing regimens (HR = 1.86, 95 %-CI 1.33-2.61) and filgrastim (HR = 1.47, 95 %-CI 1.05-2.06), but not pegfilgrastim (HR = 1.10, 95 %-CI 0.73-1.66). We observed increased MDS/AML risk among older breast cancer survivors treated with anthracycline/cyclophosphamide chemotherapy that was enhanced by G-CSF. Although small, this risk warrants consideration when determining adjuvant chemotherapy and neutropenia prophylaxis for breast cancer patients.
Collapse
Affiliation(s)
- Gregory S Calip
- Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago, 833 South Wood Street M/C 871, Chicago, IL, 60612-7230, USA.
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| | - Judith A Malmgren
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- HealthStat Consulting, Inc., Seattle, WA, USA
| | - Wan-Ju Lee
- Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago, 833 South Wood Street M/C 871, Chicago, IL, 60612-7230, USA
| | - Stephen M Schwartz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
139
|
Katakura F, Yabu T, Yamaguchi T, Miyamae J, Shirinashihama Y, Nakanishi T, Moritomo T. Exploring erythropoiesis of common carp (Cyprinus carpio) using an in vitro colony assay in the presence of recombinant carp kit ligand A and erythropoietin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 53:13-22. [PMID: 26111997 DOI: 10.1016/j.dci.2015.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/11/2015] [Accepted: 06/11/2015] [Indexed: 06/04/2023]
Abstract
The use of in vitro colony assays in mammals has contributed to identification of erythroid progenitor cells such as burst-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E) progenitors, and serves to examine functions of erythropoietic growth factors like Erythropoietin (Epo) and Kit ligand. Here, we established an in vitro colony-forming assay capable of investigating erythropoiesis in carp (Cyprinus carpio), cloned and functionally characterized recombinant homologous molecules Epo and Kit ligand A (Kitla), and identified three distinct erythroid progenitor cells in carp. Recombinant carp Epo induced the formation of CFU-E-like and BFU-E-like erythroid colonies, expressing erythroid marker genes, β-globin, epor and gata1. Recombinant carp Kitla alone induced limited colony formation, whereas a combination of Kitla and Epo dramatically enhanced erythroid colony formation and colony cell growth, as well as stimulated the formation of thrombocytic/erythroid colonies expressing not only erythroid markers but also thrombocytic markers, cd41 and c-mpl. Utilizing this colony assay to examine the distribution of distinct erythroid progenitor cells in carp, we demonstrated that carp head and trunk kidney play a primary role in erythropoiesis, while the spleen plays a secondary. Furthermore, we showed that presumably bi-potent thrombocytic/erythroid progenitor cells localize principally in the trunk kidney. Our results indicate that teleost fish possess mechanisms of Epo- and Kitla-dependent erythropoiesis similar to those in other vertebrates, and also help to demonstrate the diversity of erythropoietic sites among vertebrates.
Collapse
Affiliation(s)
- Fumihiko Katakura
- Laboratory of Comparative Immunology, Department of Veterinary Medicine, Nihon University, Kameino 1866, Fujisawa, Kanagawa 252-0880, Japan.
| | - Takeshi Yabu
- Laboratory of Fish Pathology, Department of Veterinary Medicine, Nihon University, Japan
| | - Takuya Yamaguchi
- Laboratory of Fish Pathology, Department of Veterinary Medicine, Nihon University, Japan
| | - Jiro Miyamae
- Laboratory of Comparative Immunology, Department of Veterinary Medicine, Nihon University, Kameino 1866, Fujisawa, Kanagawa 252-0880, Japan
| | - Yuki Shirinashihama
- Laboratory of Fish Pathology, Department of Veterinary Medicine, Nihon University, Japan
| | - Teruyuki Nakanishi
- Laboratory of Fish Pathology, Department of Veterinary Medicine, Nihon University, Japan
| | - Tadaaki Moritomo
- Laboratory of Comparative Immunology, Department of Veterinary Medicine, Nihon University, Kameino 1866, Fujisawa, Kanagawa 252-0880, Japan
| |
Collapse
|
140
|
Origins of the Vertebrate Erythro/Megakaryocytic System. BIOMED RESEARCH INTERNATIONAL 2015; 2015:632171. [PMID: 26557683 PMCID: PMC4628740 DOI: 10.1155/2015/632171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/02/2015] [Indexed: 02/08/2023]
Abstract
Vertebrate erythrocytes and thrombocytes arise from the common bipotent thrombocytic-erythroid progenitors (TEPs). Even though nonmammalian erythrocytes and thrombocytes are phenotypically very similar to each other, mammalian species have developed some key evolutionary improvements in the process of erythroid and thrombocytic differentiation, such as erythroid enucleation, megakaryocyte endoreduplication, and platelet formation. This brings up a few questions that we try to address in this review. Specifically, we describe the ontology of erythro-thrombopoiesis during adult hematopoiesis with focus on the phylogenetic origin of mammalian erythrocytes and thrombocytes (also termed platelets). Although the evolutionary relationship between mammalian and nonmammalian erythroid cells is clear, the appearance of mammalian megakaryocytes is less so. Here, we discuss recent data indicating that nonmammalian thrombocytes and megakaryocytes are homologs. Finally, we hypothesize that erythroid and thrombocytic differentiation evolved from a single ancestral lineage, which would explain the striking similarities between these cells.
Collapse
|
141
|
Payuhakrit W, Panichakul T, Charoenphon N, Chalermsaenyakorn P, Jaovisidha A, Wongborisuth C, Udomsangpetch R. In vitro production of functional immune cells derived from human hematopoietic stem cells. EXCLI JOURNAL 2015; 14:1031-9. [PMID: 26933404 PMCID: PMC4763471 DOI: 10.17179/excli2015-506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/01/2015] [Indexed: 01/18/2023]
Abstract
Hematopoietic stem cells (HSC) from cord blood are potentially high sources for transplantation due to their low immunogenicity and the presence of the multipotent cells. These cells are capable of differentiating to produce various lineages of blood cells under specific conditions. We have enriched highly purified CD34+ cells from cord blood, determined in vitro growth of the cells in culture systems in the absence (condition A) or presence of GM-CSF and G-CSF (condition B), and determined the profile of immune cells during the period of cultivation by using flow cytometry. PhytohemagglutininA (PHA) was used as a mitogen to stimulate T lymphocytes derived from hematopoietic stem cells. GM-CSF and G-CSF prolonged the survival of the growing cells and also maintained expansion of cells in blastic stage. By day 12 of cultivation, when cell numbers peaked, various types of immune cells had appeared (CD14+ cells, CD40+HLA-DR+ cells, CD3+CD56+ cells, CD19+ cells, CD3+CD4+ cells, CD3+CD8+cells and CD3-CD56+). A significantly higher percentage of monocytes (p = 0.002) were observed under culture with GM-CSF, G-CSF when compared with culture without GM-CSF, G-CSF. In addition, T lymphocytes derived from HSC responded to 50 µg/ml of PHA. This is the first report showing the complete differentiation and proliferation of immune cells derived from CD34+ HSC under in vitro culture conditions. Lymphocytes, monocytes, dendritic cells and polymorph nuclear cells derived from HSC in vitro are unique, and thus may benefit various studies such as innate immunity and pathophysiology of immune disorders.
Collapse
Affiliation(s)
- Witchuda Payuhakrit
- Department of Pathobiology, Faculty of Science, Mahidol University, Thailand
| | - Tasanee Panichakul
- Faculty of Science and Technology, Suan Dusit Rajabhat University, Thailand
| | | | | | - Adithep Jaovisidha
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Chokdee Wongborisuth
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Rachanee Udomsangpetch
- Department of Pathobiology, Faculty of Science, Mahidol University, Thailand; Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Thailand; Center for Emerging and Neglected Infectious Diseases, Mahidol University, Thailand
| |
Collapse
|
142
|
Frydman GH, Davis N, Beck PL, Fox JG. Helicobacter pylori Eradication in Patients with Immune Thrombocytopenic Purpura: A Review and the Role of Biogeography. Helicobacter 2015; 20:239-51. [PMID: 25728540 PMCID: PMC4506733 DOI: 10.1111/hel.12200] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Idiopathic thrombocytopenic purpura (ITP) is typically a diagnosis of exclusion, assigned by clinicians after ruling out other identifiable etiologies. Since a report by Gasbarrini et al. in 1998, an accumulating body of evidence has proposed a pathophysiological link between ITP and chronic Helicobacter pylori (H. pylori) infection. Clinical reports have described a spontaneous resolution of ITP symptoms in about 50% of chronic ITP patients following empirical treatment of H. pylori infection, but response appears to be geography dependent. Studies have also documented that ITP patients in East Asian countries are more likely to express positive antibody titers against H. pylori-specific cytotoxic-associated gene A (CagA), a virulence factor that is associated with an increased risk for gastric diseases including carcinoma. While a definitive mechanism by which H. pylori may induce thrombocytopenia remains elusive, proposed pathways include molecular mimicry of CagA by host autoantibodies against platelet surface glycoproteins, as well as perturbations in the phagocytic activity of monocytes. Traditional treatments of ITP have been largely empirical, involving the use of immunosuppressive agents and immunoglobulin therapy. However, based on the findings of clinical reports emerging over the past 20 years, health organizations around the world increasingly suggest the detection and eradication of H. pylori as a treatment for ITP. Elucidating the exact molecular mechanisms of platelet activation in H. pylori-positive ITP patients, while considering biogeographical differences in response rates, could offer insight into how best to use clinical H. pylori eradication to treat ITP, but will require well-designed studies to confirm the suggested causative relationship between bacterial infection and an autoimmune disease state.
Collapse
Affiliation(s)
- Galit H. Frydman
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nick Davis
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paul L. Beck
- The Gastrointestinal Research Group, Division of Gastroenterology, University of Calgary, Calgary, AB, Canada
| | - James G. Fox
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
143
|
Bernlochner I, Goedel A, Plischke C, Schüpke S, Haller B, Schulz C, Mayer K, Morath T, Braun S, Schunkert H, Siess W, Kastrati A, Laugwitz KL. Impact of immature platelets on platelet response to ticagrelor and prasugrel in patients with acute coronary syndrome. Eur Heart J 2015. [DOI: 10.1093/eurheartj/ehv326] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
144
|
Trinh BQ, Barengo N, Kim SB, Lee JS, Zweidler-McKay PA, Naora H. The homeobox gene DLX4 regulates erythro-megakaryocytic differentiation by stimulating IL-1β and NF-κB signaling. J Cell Sci 2015. [PMID: 26208636 PMCID: PMC4541043 DOI: 10.1242/jcs.168187] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Megakaryocyte and erythroid development are tightly controlled by a repertoire of cytokines, but it is not clear how cytokine-activated signaling pathways are controlled during development of these two lineages. Here, we identify that expression of DLX4, a transcription factor encoded by a homeobox gene, increases during megakaryopoiesis but decreases during erythropoiesis. Enforced expression of DLX4 in CD34(+) stem and progenitor cells and in bipotent K562 cells induced lineage markers and morphologic features of megakaryocytes and repressed erythroid marker expression and hemoglobin levels. Converse results were obtained when DLX4 was knocked down. Gene Ontology and Gene Set Enrichment Analyses of genome-wide changes in gene expression revealed that DLX4 induces a megakaryocytic transcriptional program and inhibits an erythroid transcriptional program. DLX4 also induced gene signatures that are associated with nuclear factor κB (NF-κB) signaling. The ability of DLX4 to promote megakaryocyte development at the expense of erythroid generation was diminished by blocking NF-κB activity or by repressing IL1B, a transcriptional target of DLX4. Collectively, our findings indicate that DLX4 exerts opposing effects on the megakaryocytic and erythroid lineages in part by inducing IL-1β and NF-κB signaling.
Collapse
Affiliation(s)
- Bon Q Trinh
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 108, Houston, TX 77030, USA
| | - Nicolas Barengo
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 108, Houston, TX 77030, USA
| | - Sang Bae Kim
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 950, Houston, TX 77030, USA
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 950, Houston, TX 77030, USA
| | - Patrick A Zweidler-McKay
- Division of Pediatrics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 853, Houston, TX 77030, USA
| | - Honami Naora
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 108, Houston, TX 77030, USA
| |
Collapse
|
145
|
Kazanis I, Feichtner M, Lange S, Rotheneichner P, Hainzl S, Öller M, Schallmoser K, Rohde E, Reitsamer HA, Couillard-Despres S, Bauer HC, Franklin RJM, Aigner L, Rivera FJ. Lesion-induced accumulation of platelets promotes survival of adult neural stem / progenitor cells. Exp Neurol 2015; 269:75-89. [PMID: 25819103 DOI: 10.1016/j.expneurol.2015.03.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/21/2015] [Accepted: 03/20/2015] [Indexed: 12/22/2022]
Abstract
The presence of neural stem/progenitor cells (NSPCs) in specific areas of the central nervous system (CNS) supports tissue maintenance as well as regeneration. The subependymal zone (SEZ), located at the lateral ventricle's wall, represents a niche for NSPCs and in response to stroke or demyelination becomes activated with progenitors migrating towards the lesion and differentiating into neurons and glia. The mechanisms that underlie this phenomenon remain largely unknown. The vascular niche and in particular blood-derived elements such as platelets, has been shown to contribute to CNS regeneration in different pathological conditions. Indeed, intracerebroventricularly administrated platelet lysate (PL) stimulates angiogenesis, neurogenesis and neuroprotection in the damaged CNS. Here, we explored the presence of platelets in the activated SEZ after a focal demyelinating lesion in the corpus callosum of mice and we studied the effects of PL on proliferating SEZ-derived NSPCs in vitro. We showed that the lesion-induced increase in the size of the SEZ and in the number of proliferating SEZ-resident NSPCs correlates with the accumulation of platelets specifically along the activated SEZ vasculature. Expanding on this finding, we demonstrated that exposure of NSPCs to PL in vitro led to increased numbers of cells by enhanced cell survival and reduced apoptosis without differences in proliferation and in the differentiation potential of NSPCs. Finally, we demonstrate that the accumulation of platelets within the SEZ is spatially correlated with reduced numbers of apoptotic cells when compared to other periventricular areas. In conclusion, our results show that platelet-derived compounds specifically promote SEZ-derived NSPC survival and suggest that platelets might contribute to the enlargement of the pool of SEZ NSPCs that are available for CNS repair in response to injury.
Collapse
Affiliation(s)
- Ilias Kazanis
- Wellcome Trust and MRC Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Biology, University of Patras, Patras, Greece
| | - Martina Feichtner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Department of Blood Group Serology and Transfusion Medicine, Federal Hospital and Paracelsus Medical University Salzburg Salzburg, Salzburg, Austria
| | - Simona Lange
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Peter Rotheneichner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Institute of Experimental Neuroregeneration, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Stefan Hainzl
- Division of Molecular Dermatology and EB House Austria, Department of Dermatology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Michaela Öller
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Department of Blood Group Serology and Transfusion Medicine, Federal Hospital and Paracelsus Medical University Salzburg Salzburg, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Department of Blood Group Serology and Transfusion Medicine, Federal Hospital and Paracelsus Medical University Salzburg Salzburg, Salzburg, Austria
| | - Eva Rohde
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Department of Blood Group Serology and Transfusion Medicine, Federal Hospital and Paracelsus Medical University Salzburg Salzburg, Salzburg, Austria
| | - Herbert A Reitsamer
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Ophthalmology/Optometry and Research Program for Experimental Ophthalmology, Paracelsus Medical University, Salzburg, Austria
| | - Sebastien Couillard-Despres
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Institute of Experimental Neuroregeneration, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Hans-Christian Bauer
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Institute of Tendon and Bone Regeneration, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Robin J M Franklin
- Wellcome Trust and MRC Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Francisco J Rivera
- Wellcome Trust and MRC Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom; Institute of Molecular Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria.
| |
Collapse
|
146
|
Bhattacharya P, Thiruppathi M, Elshabrawy HA, Alharshawi K, Kumar P, Prabhakar BS. GM-CSF: An immune modulatory cytokine that can suppress autoimmunity. Cytokine 2015; 75:261-71. [PMID: 26113402 DOI: 10.1016/j.cyto.2015.05.030] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/22/2015] [Accepted: 05/25/2015] [Indexed: 12/12/2022]
Abstract
GM-CSF was originally identified as a colony stimulating factor (CSF) because of its ability to induce granulocyte and macrophage populations from precursor cells. Multiple studies have demonstrated that GM-CSF is also an immune-modulatory cytokine, capable of affecting not only the phenotype of myeloid lineage cells, but also T-cell activation through various myeloid intermediaries. This property has been implicated in the sustenance of several autoimmune diseases like arthritis and multiple sclerosis. In contrast, several studies using animal models have shown that GM-CSF is also capable of suppressing many autoimmune diseases such as Crohn's disease, Type-1 diabetes, Myasthenia gravis and experimental autoimmune thyroiditis. Knockout mouse studies have suggested that the role of GM-CSF in maintaining granulocyte and macrophage populations in the physiological steady state is largely redundant. Instead, its immune-modulatory role plays a significant role in the development or resolution of autoimmune diseases. This is mediated either through the differentiation of precursor cells into specialized non-steady state granulocytes, macrophages and dendritic cells, or through the modulation of the phenotype of mature myeloid cells. Thus, outside of myelopoiesis, GM-CSF has a profound role in regulating the immune response and maintaining immunological tolerance.
Collapse
Affiliation(s)
- Palash Bhattacharya
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Muthusamy Thiruppathi
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Hatem A Elshabrawy
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Khaled Alharshawi
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
147
|
Schumacher A, Denecke B, Braunschweig T, Stahlschmidt J, Ziegler S, Brandenburg LO, Stope MB, Martincuks A, Vogt M, Görtz D, Camporeale A, Poli V, Müller-Newen G, Brümmendorf TH, Ziegler P. Angptl4 is upregulated under inflammatory conditions in the bone marrow of mice, expands myeloid progenitors, and accelerates reconstitution of platelets after myelosuppressive therapy. J Hematol Oncol 2015; 8:64. [PMID: 26054961 PMCID: PMC4460974 DOI: 10.1186/s13045-015-0152-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/07/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Upon inflammation, myeloid cell generation in the bone marrow (BM) is broadly enhanced by the action of induced cytokines which are produced locally and at multiple sites throughout the body. METHODS Using microarray studies, we found that Angptl4 is upregulated in the BM during systemic inflammation. RESULTS Recombinant murine Angptl4 (rmAngptl4) stimulated the proliferation of myeloid colony-forming units (CFUs) in vitro. Upon repeated in vivo injections, rmAngptl4 increased BM progenitor cell frequency and this was paralleled by a relative increase in phenotypically defined granulocyte-macrophage progenitors (GMPs). Furthermore, in vivo treatment with rmAngptl4 resulted in elevated platelet counts in steady-state mice while allowing a significant acceleration of reconstitution of platelets after myelosuppressive therapy. The administration of rmAngptl4 increased the number of CD61(+)CD41(low)-expressing megakaryocytes (MK) in the BM of steady-state and in the spleen of transplanted mice. Furthermore, rmAngptl4 improved the in vitro differentiation of immature MKs from hematopoietic stem and progenitor cells. Mechanistically, using a signal transducer and activator of transcription 3 (STAT3) reporter knockin model, we show that rmAngptl4 induces de novo STAT3 expression in immature MK which could be important for the effective expansion of MKs after myelosuppressive therapy. CONCLUSION Whereas the definitive role of Angptl4 in mediating the effects of lipopolysaccharide (LPS) on the BM has to be demonstrated by further studies involving multiple cytokine knockouts, our data suggest that Angptl4 plays a critical role during hematopoietic, especially megakaryopoietic, reconstitution following stem cell transplantation.
Collapse
Affiliation(s)
- Anne Schumacher
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research IZKF Aachen, RWTH Aachen University Hospital, Aachen, Germany.
| | - Till Braunschweig
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Jasmin Stahlschmidt
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Susanne Ziegler
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany.
| | - Antons Martincuks
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Michael Vogt
- Institute for Laboratory Animal Science, University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Dieter Görtz
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Annalisa Camporeale
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10126, Turin, Italy.
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10126, Turin, Italy.
| | - Gerhard Müller-Newen
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Tim H Brümmendorf
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Patrick Ziegler
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute for Occupational and Social Medicine, Aachen University, Aachen, Germany.
| |
Collapse
|
148
|
Thrombocytopenia in Dengue: Interrelationship between Virus and the Imbalance between Coagulation and Fibrinolysis and Inflammatory Mediators. Mediators Inflamm 2015; 2015:313842. [PMID: 25999666 PMCID: PMC4427128 DOI: 10.1155/2015/313842] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 01/22/2015] [Indexed: 01/15/2023] Open
Abstract
Dengue is an infectious disease caused by dengue virus (DENV). In general, dengue is a self-limiting acute febrile illness followed by a phase of critical defervescence, in which patients may improve or progress to a severe form. Severe illness is characterized by hemodynamic disturbances, increased vascular permeability, hypovolemia, hypotension, and shock. Thrombocytopenia and platelet dysfunction are common in both cases and are related to the clinical outcome. Different mechanisms have been hypothesized to explain DENV-associated thrombocytopenia, including the suppression of bone marrow and the peripheral destruction of platelets. Studies have shown DENV-infected hematopoietic progenitors or bone marrow stromal cells. Moreover, anti-platelet antibodies would be involved in peripheral platelet destruction as platelets interact with endothelial cells, immune cells, and/or DENV. It is not yet clear whether platelets play a role in the viral spread. Here, we focus on the mechanisms of thrombocytopenia and platelet dysfunction in DENV infection. Because platelets participate in the inflammatory and immune response by promoting cytokine, chemokine, and inflammatory mediator secretion, their relevance as "immune-like effector cells" will be discussed. Finally, an implication for platelets in plasma leakage will be also regarded, as thrombocytopenia is associated with clinical outcome and higher mortality.
Collapse
|
149
|
Gordan R, Gwathmey JK, Xie LH. Autonomic and endocrine control of cardiovascular function. World J Cardiol 2015; 7:204-214. [PMID: 25914789 PMCID: PMC4404375 DOI: 10.4330/wjc.v7.i4.204] [Citation(s) in RCA: 384] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 01/22/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023] Open
Abstract
The function of the heart is to contract and pump oxygenated blood to the body and deoxygenated blood to the lungs. To achieve this goal, a normal human heart must beat regularly and continuously for one’s entire life. Heartbeats originate from the rhythmic pacing discharge from the sinoatrial (SA) node within the heart itself. In the absence of extrinsic neural or hormonal influences, the SA node pacing rate would be about 100 beats per minute. Heart rate and cardiac output, however, must vary in response to the needs of the body’s cells for oxygen and nutrients under varying conditions. In order to respond rapidly to the changing requirements of the body’s tissues, the heart rate and contractility are regulated by the nervous system, hormones, and other factors. Here we review how the cardiovascular system is controlled and influenced by not only a unique intrinsic system, but is also heavily influenced by the autonomic nervous system as well as the endocrine system.
Collapse
|
150
|
Pineault N, Boisjoli GJ. Megakaryopoiesis andex vivodifferentiation of stem cells into megakaryocytes and platelets. ACTA ACUST UNITED AC 2015. [DOI: 10.1111/voxs.12155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- N. Pineault
- Center for Innovation; Canadian Blood Services; Ottawa ON Canada
- Department of Biochemistry, Microbiology and Immunology; University of Ottawa; Ottawa ON Canada
| | - G. J. Boisjoli
- Center for Innovation; Canadian Blood Services; Ottawa ON Canada
| |
Collapse
|