1
|
Burtscher J, Citherlet T, Camacho-Cardenosa A, Camacho-Cardenosa M, Raberin A, Krumm B, Hohenauer E, Egg M, Lichtblau M, Müller J, Rybnikova EA, Gatterer H, Debevec T, Baillieul S, Manferdelli G, Behrendt T, Schega L, Ehrenreich H, Millet GP, Gassmann M, Schwarzer C, Glazachev O, Girard O, Lalande S, Hamlin M, Samaja M, Hüfner K, Burtscher M, Panza G, Mallet RT. Mechanisms underlying the health benefits of intermittent hypoxia conditioning. J Physiol 2024; 602:5757-5783. [PMID: 37860950 DOI: 10.1113/jp285230] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Intermittent hypoxia (IH) is commonly associated with pathological conditions, particularly obstructive sleep apnoea. However, IH is also increasingly used to enhance health and performance and is emerging as a potent non-pharmacological intervention against numerous diseases. Whether IH is detrimental or beneficial for health is largely determined by the intensity, duration, number and frequency of the hypoxic exposures and by the specific responses they engender. Adaptive responses to hypoxia protect from future hypoxic or ischaemic insults, improve cellular resilience and functions, and boost mental and physical performance. The cellular and systemic mechanisms producing these benefits are highly complex, and the failure of different components can shift long-term adaptation to maladaptation and the development of pathologies. Rather than discussing in detail the well-characterized individual responses and adaptations to IH, we here aim to summarize and integrate hypoxia-activated mechanisms into a holistic picture of the body's adaptive responses to hypoxia and specifically IH, and demonstrate how these mechanisms might be mobilized for their health benefits while minimizing the risks of hypoxia exposure.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Tom Citherlet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Alba Camacho-Cardenosa
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Marta Camacho-Cardenosa
- Clinical Management Unit of Endocrinology and Nutrition - GC17, Maimónides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofía University Hospital, Córdoba, Spain
| | - Antoine Raberin
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Bastien Krumm
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Erich Hohenauer
- Rehabilitation and Exercise Science Laboratory (RES lab), Department of Business Economics, Health and Social Care, University of Applied Sciences and Arts of Southern Switzerland, Landquart, Switzerland
- International University of Applied Sciences THIM, Landquart, Switzerland
- Department of Neurosciences and Movement Science, University of Fribourg, Fribourg, Switzerland
| | - Margit Egg
- Institute of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Mona Lichtblau
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Julian Müller
- Department of Pulmonology, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Elena A Rybnikova
- Pavlov Institute of Physiology, Russian Academy of Sciences, St Petersburg, Russia
| | - Hannes Gatterer
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
- Institute for Sports Medicine, Alpine Medicine and Health Tourism (ISAG), UMIT TIROL-Private University for Health Sciences and Health Technology, Hall in Tirol, Austria
| | - Tadej Debevec
- Faculty of Sport, University of Ljubljana, Ljubljana, Slovenia
- Department of Automatics, Biocybernetics and Robotics, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Sebastien Baillieul
- Service Universitaire de Pneumologie Physiologie, University of Grenoble Alpes, Inserm, Grenoble, France
| | | | - Tom Behrendt
- Chair Health and Physical Activity, Department of Sport Science, Institute III, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Lutz Schega
- Chair Health and Physical Activity, Department of Sport Science, Institute III, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Hannelore Ehrenreich
- Clinical Neuroscience, University Medical Center and Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Christoph Schwarzer
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Oleg Glazachev
- Department of Normal Physiology, N.V. Sklifosovsky Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olivier Girard
- School of Human Sciences (Exercise and Sport Science), The University of Western Australia, Crawley, Western Australia, Australia
| | - Sophie Lalande
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, TX, USA
| | - Michael Hamlin
- Department of Tourism, Sport and Society, Lincoln University, Christchurch, New Zealand
| | - Michele Samaja
- Department of Health Science, University of Milan, Milan, Italy
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Hospital for Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Gino Panza
- The Department of Health Care Sciences, Program of Occupational Therapy, Wayne State University, Detroit, MI, USA
- John D. Dingell VA Medical Center Detroit, Detroit, MI, USA
| | - Robert T Mallet
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
2
|
Chevrier A, Lavertu M. Manufacturing Process Affects Coagulation Kinetics of Ortho-R, an Injectable Chitosan-Platelet-Rich Plasma Biomaterial for Tissue Repair. Bioengineering (Basel) 2024; 11:929. [PMID: 39329671 PMCID: PMC11428962 DOI: 10.3390/bioengineering11090929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Ortho-R (ChitogenX Inc., Kirkland, QC, Canada) is an injectable combination drug-biologic product that is used as an adjunct to augment the standard of care for the surgical repair of soft tissues. The drug product comprises lyophilized chitosan, trehalose and calcium chloride, and it is dissolved in platelet-rich plasma (PRP), a blood-derived biologic, prior to injection at the surgical site where it will coagulate. The first step of the Ortho-R manufacturing process involves dissolving the chitosan in hydrochloric acid. The purpose of this study was to investigate the effect of increasing the amount of acid used to dissolve the chitosan on final drug product performance, more specifically, on the chitosan-PRP coagulation kinetics. Chitosans were solubilized in hydrochloric acid, with concentrations adjusted to obtain between 60% and 95% protonation of the chitosan amino groups. Freeze-dried Ortho-R was solubilized with PRP, and coagulation was assessed using thromboelastography (TEG). The clotted mixtures were observed with histology. Clot reaction time (TEG R) increased and clot maximal amplitude (TEG MA) decreased with protonation levels as pH decreased. Chitosan distribution was homogeneous in chitosan-PRP clots at the lowest protonation levels, but it accumulated toward the surface of the clots at the highest protonation levels as pH decreased. These changes in coagulation kinetics, clot strength and chitosan distribution induced by high protonation of the chitosan amino groups were partially reversed by adding sodium hydroxide to the dissolved chitosan component in order to decrease pH. Careful control of manufacturing processes is critical, and it is important to consider the impact of each manufacturing step on product performance.
Collapse
Affiliation(s)
- Anik Chevrier
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
3
|
Kim Y, Bae H, Yu D. The in vitro effects of acidemia and acidemia reversal on coagulation in dogs. Front Vet Sci 2024; 11:1427237. [PMID: 39301280 PMCID: PMC11410758 DOI: 10.3389/fvets.2024.1427237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Background The effect of acidemia on blood coagulation remains inadequately understood in veterinary medicine. Therefore, we assessed the effect of in vitro acidification of canine whole blood on coagulation and investigated whether acidemia-induced coagulopathy could be reversed by reversing acidemia. Methods Citrated whole blood samples were taken from six healthy Beagle dogs and categorized, based on pH adjustment, into neutral, weak acidemia (WA), strong acidemia (SA), and reversal from SA. Then, prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen concentration, conventional thromboelastography (TEG) parameters, and velocity curve (V-curve) variables of TEG were assessed. Results The PT, aPTT, and most TEG parameters showed significant coagulopathy in the SA group compared to the neutral group, with additional significant changes in reaction time (R), clot kinetic (K), maximum amplitude (MA), split point (SP), elasticity (E), thrombodynamic potential index (TPI), and coagulation index (CI) between the SA and WA groups. Among V-curve variables, the maximum rate of thrombus generation (MRTG) and total thrombus generation were significantly inhibited in the SA group compared to the neutral group, with significant differences in the time to maximum rate of thrombus generation (TMRTG) between the WA and SA groups. In the reverse group, aPTT, R, K, α-angle, MRTG, TMRTG, SP, TPI, and CI exhibited significant recovery compared to the SA group. Conclusion The in vitro induction of acidemia in canine whole blood leads to impairment of coagulation profiles, and pH correction can reverse most acidemia-induced coagulopathy.
Collapse
Affiliation(s)
- Youngju Kim
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyeona Bae
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - DoHyeon Yu
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
4
|
Ledergerber K, Hollinger A, Zimmermann S, Todorov A, Trutmann M, Gallachi L, Gschwandtner LA, Ryser LA, Gebhard CE, Bolliger D, Buser A, Tsakiris DA, Siegemund M. Impact of Additional Administration of von Willebrand Factor Concentrates to Thrombocyte Transfusion in Perioperative Bleeding in Cardiac Surgery. Transfus Med Hemother 2024; 51:22-31. [PMID: 38314243 PMCID: PMC10836859 DOI: 10.1159/000530810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/16/2023] [Indexed: 02/06/2024] Open
Abstract
Background Von Willebrand factor (vWF) is an important part of blood coagulation since it binds platelets to each other and to endothelial cells. In traumatic and surgical haemorrhage, both blood cells and plasmatic factors are consumed, leading to consumption coagulopathy and fluid resuscitation. This often results in large amounts of crystalloids and blood products being infused. Additional administration of vWF complex and platelets might mitigate this problem. We hypothesize that administration of vWF concentrate additionally to platelet concentrates reduces blood loss and the amount of blood products (platelets, red blood cells [RBC], fresh frozen plasma [FFP]) administered. Methods We conducted a monocentric 6-year retrospective data analysis of cardiac surgery patients. Included were all patients receiving platelet concentrates within 48 h postoperatively. Patients who additionally received vWF concentrates were allocated to the intervention group and all others to the control group. Groups were compared in mixed regression models correcting for known confounders, based on nearest neighbour propensity score matching. Primary endpoints were loss of blood (day one and two) and amount of needed blood products on day one and two (platelets, RBC, FFP). Secondary endpoints were intensive care unit (ICU) and in-hospital length of stay, ICU and in-hospital mortality, and absolute difference of platelet counts before and after treatment. Results Of 497 patients analysed, 168 (34%) received vWF concentrates. 121 patients in both groups were considered for nearest neighbour matching. Patients receiving additional vWF were more likely to receive more blood products (RBC, FFP, platelets) in the first 24 h after surgery and had around 200 mL more blood loss at the same time. Conclusion In this retrospective analysis, no benefit in additional administration of vWF to platelet concentrates on perioperative blood loss, transfusion requirement (platelets, RBC, FFP), length of stay, and mortality could be found. These findings should be verified in a prospective randomized controlled clinical trial (www.clinicaltrials.gov identifier NCT04555785).
Collapse
Affiliation(s)
| | - Alexa Hollinger
- Intensive Care Unit, University Hospital Basel, Basel, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
| | | | - Atanas Todorov
- Cardiovascular Gender Medicine, University Hospital Zürich, Zürich, Switzerland
| | - Maren Trutmann
- Intensive Care Unit, University Hospital Basel, Basel, Switzerland
| | - Laura Gallachi
- Intensive Care Unit, University Hospital Basel, Basel, Switzerland
| | | | | | - Caroline Eva Gebhard
- Intensive Care Unit, University Hospital Basel, Basel, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
| | - Daniel Bolliger
- Medical Faculty, University of Basel, Basel, Switzerland
- Department of Anesthesiology, University Hospital Basel, Basel, Switzerland
| | - Andreas Buser
- Medical Faculty, University of Basel, Basel, Switzerland
- Department of Anesthesiology, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service of the Swiss Red Cross, Basel, Switzerland
| | - Dimitrios Athanasios Tsakiris
- Medical Faculty, University of Basel, Basel, Switzerland
- Department of Transfusion Medicine and Hematology, Basel University Hospital, Basel, Switzerland
| | - Martin Siegemund
- Intensive Care Unit, University Hospital Basel, Basel, Switzerland
- Medical Faculty, University of Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Hong C, He Y, Bowen PA, Belcher AM, Olsen BD, Hammond PT. Engineering a Two-Component Hemostat for the Treatment of Internal Bleeding through Wound-Targeted Crosslinking. Adv Healthc Mater 2023; 12:e2202756. [PMID: 37017403 PMCID: PMC10964210 DOI: 10.1002/adhm.202202756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/01/2023] [Indexed: 04/06/2023]
Abstract
Primary hemostasis (platelet plug formation) and secondary hemostasis (fibrin clot formation) are intertwined processes that occur upon vascular injury. Researchers have sought to target wounds by leveraging cues specific to these processes, such as using peptides that bind activated platelets or fibrin. While these materials have shown success in various injury models, they are commonly designed for the purpose of treating solely primary or secondary hemostasis. In this work, a two-component system consisting of a targeting component (azide/GRGDS PEG-PLGA nanoparticles) and a crosslinking component (multifunctional DBCO) is developed to treat internal bleeding. The system leverages increased injury accumulation to achieve crosslinking above a critical concentration, addressing both primary and secondary hemostasis by amplifying platelet recruitment and mitigating plasminolysis for greater clot stability. Nanoparticle aggregation is measured to validate concentration-dependent crosslinking, while a 1:3 azide/GRGDS ratio is found to increase platelet recruitment, decrease clot degradation in hemodiluted environments, and decrease complement activation. Finally, this approach significantly increases survival relative to the particle-only control in a liver resection model. In light of prior successes with the particle-only system, these results emphasize the potential of this technology in aiding hemostasis and the importance of a holistic approach in engineering new treatments for hemorrhage.
Collapse
Affiliation(s)
- Celestine Hong
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Yanpu He
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Porter A. Bowen
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Angela M. Belcher
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Bradley D. Olsen
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Paula T. Hammond
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
6
|
Ahmad R, Narwaria M, Singh A, Kumar S, Haque M. Detecting Diabetic Ketoacidosis with Infection: Combating a Life-Threatening Emergency with Practical Diagnostic Tools. Diagnostics (Basel) 2023; 13:2441. [PMID: 37510185 PMCID: PMC10378387 DOI: 10.3390/diagnostics13142441] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Diabetic ketoacidosis (DKA) is a life-threatening acute complication of diabetes mellitus and can lead to patient demise if not immediately treated. From the recent literature, the diabetic ketoacidosis mortality rate, depending on age, is 2-5%. Insulin discontinuation and infection remain the two most common triggers for diabetic ketoacidosis. About 50% of cases of ketoacidosis result from bacterial infections like urinary tract infections and pneumonia. It is also important to diagnose the presence of infection in diabetic ketoacidosis patients to prevent the excessive use of antibiotics, which may lead to antibiotic resistance. Although performing bacterial culture is confirmatory for the presence or absence of bacterial infection, the time required to obtain the result is long. At the same time, emergency treatment needs to be started as early as possible. METHODS This narrative review examines various septic markers to identify the appropriate tools for diagnosis and to distinguish between diabetic ketoacidosis with and without infection. Electronic databases were searched using the Google engine with the keywords "Diabetes Mellitus", "Diabetic Ketoacidosis", "Infection with Diabetic Ketoacidosis", "biomarkers for infection in Diabetic Ketoacidosis", "Procalcitonin", "Inflammatory cytokines in DKA", "Lactic acidosis in DKA", and "White blood cell in infection in DKA". RESULTS This narrative review article presents the options for diagnosis and also aims to create awareness regarding the gravity of diabetic ketoacidosis with infection and emphasizes the importance of early diagnosis for appropriate management. Diabetes mellitus is a clinical condition that may lead to several acute and chronic complications. Acute diabetic ketoacidosis is a life-threatening condition in which an excess production of ketone bodies results in acidosis and hypovolemia. Infection is one of the most common triggers of diabetic ketoacidosis. When bacterial infection is present along with diabetic ketoacidosis, the mortality rate is even higher than for patients with diabetic ketoacidosis without infection. The symptoms and biomarkers of diabetic ketoacidosis are similar to that of infection, like fever, C reactive protein, and white blood cell count, since both create an environment of systemic inflammation. It is also essential to distinguish between the presence and absence of bacterial infection to ensure the appropriate use of antibiotics and prevent antimicrobial resistance. A bacterial culture report is confirmatory for the existence of bacterial infection, but this may take up to 24 h. Diagnosis needs to be performed approximately in the emergency room upon admission since there is a need for immediate management. Therefore, researching the possible diagnostic tools for the presence of infection in diabetic ketoacidosis patients is of great importance. Several of such biomarkers have been discussed in this research work.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka 1230, Bangladesh
| | - Mahendra Narwaria
- Asian Bariatrics Plus Hospital, V Wing-Mondeal Business Park, S G Highways, Ahmedabad 380054, India
| | - Arya Singh
- Asian Bariatrics Plus Hospital, V Wing-Mondeal Business Park, S G Highways, Ahmedabad 380054, India
| | - Santosh Kumar
- Department of Periodontology, Karnavati School of Dentistry, Karnavati University, Gandhinagar 382422, India
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur 57000, Malaysia
- Department of Scientific Research Center (KSRC), Karnavati School of Dentistry, Karnavati University, Gandhinagar 382422, India
| |
Collapse
|
7
|
Zhu Z, Sun S, Jiang T, Zhang L, Chen M, Chen S. A double-edged sword of platelet-derived extracellular vesicles in tissues, injury or repair: The current research overview. Tissue Cell 2023; 82:102066. [PMID: 36924675 DOI: 10.1016/j.tice.2023.102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/23/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Extracellular vesicles (EVs) are vesicular bodies with a double-layered membrane structure that are detached from the cell membrane or secreted by the cells. EVs secreted by platelets account for the main part in the blood circulation, which account for about 30% or even more. Many types of cells are regulated by PEVs, including endothelial cells, leukocytes, smooth muscle cells, etc. Nevertheless, despite the growing interest in the study of extracellular vesicles, there are still only a few studies on the role of PEVs. Therefore, this overview mainly focuses on one method of isolation and the functions of PEVs in tissues found so far, including promoting tissue repair and mediating tissue damage, which can be used for researchers to continue to explore the role of PEVs in other fields.
Collapse
Affiliation(s)
- Zepeng Zhu
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Medical School, Southeast University, Nanjing, China
| | - Si Sun
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Medical School, Southeast University, Nanjing, China
| | - Tiancheng Jiang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Medical School, Southeast University, Nanjing, China
| | - Lei Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Shuqiu Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| |
Collapse
|
8
|
Schmuckenschlager A, Pirabe A, Assinger A, Schrottmaier WC. Platelet count, temperature and pH value differentially affect hemostatic and immunomodulatory functions of platelets. Thromb Res 2023; 223:111-122. [PMID: 36738664 DOI: 10.1016/j.thromres.2023.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/29/2023]
Abstract
Platelets are primarily recognized for their role in hemostasis, but also regulate immune responses by interacting with leukocytes. Their highly sensitive nature enables platelets to rapidly respond to micro-environmental changes, which is crucial under physiological condition but can jeopardize in vitro analyses. Thus, we tested how platelet count and changes in pH and temperatures, which are commonly experienced during inflammation and infection but also affected by ex vivo analyses, influence platelet-leukocyte interaction and immunomodulation. Reducing platelet count by up to 90 % slightly decreased platelet activation and platelet-leukocyte aggregate formation, but did not affect CD11b activation nor CD62L shedding of monocytes or neutrophils. Acidosis (pH 6.9) slightly elevated platelet degranulation and binding to innate leukocytes, though pH changes did not modulate leukocyte activation. While platelet responsiveness was higher at room temperature than at 37 °C, incubation temperature did not affect platelet-leukocyte aggregate formation. In contrast, platelet-mediated CD11b activation and CD62L expression increased with temperature. Our data thus demonstrate the importance of standardized protocols for sample preparation and assay procedure to obtain comparable data. Further, unspecific physiologic responses such as thrombocytopenia, acidosis or temperature changes may contribute to platelet dysfunction and altered platelet-mediated immunomodulation in inflammatory and infectious disease.
Collapse
Affiliation(s)
- Anna Schmuckenschlager
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Waltraud C Schrottmaier
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Cartwright IM, Colgan SP. The hypoxic tissue microenvironment as a driver of mucosal inflammatory resolution. Front Immunol 2023; 14:1124774. [PMID: 36742292 PMCID: PMC9890178 DOI: 10.3389/fimmu.2023.1124774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
On the backdrop of all acute inflammatory processes lies the activation of the resolution response. Recent years have witnessed an emerging interest in defining molecular factors that influence the resolution of inflammation. A keystone feature of the mucosal inflammatory microenvironment is hypoxia. The gastrointestinal tract, particularly the colon, exists in a state of physiological hypoxia and during active inflammation, this hypoxic state is enhanced as a result of infiltrating leukocyte oxygen consumption and the activation of oxygen consuming enzymes. Most evidence suggests that mucosal hypoxia promotes the active resolution of inflammation through a variety of mechanisms, including extracellular acidification, purine biosynthesis/salvage, the generation of specialized pro-resolving lipid mediators (ie. resolvins) and altered chemokine/cytokine expression. It is now appreciated that infiltrating innate immune cells (neutrophils, eosinophils, macrophages) have an important role in molding the tissue microenvironment to program an active resolution response. Structural or functional dysregulation of this inflammatory microenvironment can result in the loss of tissue homeostasis and ultimately progression toward chronicity. In this review, we will discuss how inflammatory hypoxia drives mucosal inflammatory resolution and its impact on other microenvironmental factors that influence resolution.
Collapse
Affiliation(s)
- Ian M. Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Sean P. Colgan
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
10
|
Spiegelburg DT, Mannes M, Schultze A, Scheibenberger F, Müller F, Klitzing A, Messerer DAC, Nilsson Ekdahl K, Nilsson B, Huber-Lang M, Braun CK. Impact of surface coating and systemic anticoagulants on hemostasis and inflammation in a human whole blood model. PLoS One 2023; 18:e0280069. [PMID: 36634087 PMCID: PMC9836312 DOI: 10.1371/journal.pone.0280069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Surface compatibility with blood is critical both for scientific investigations on hemostasis and clinical applications. Regarding in vitro and ex vivo investigations, minimal alteration in physiological hemostasis is of particular importance to draw reliable conclusions on the human coagulation system. At the same time, artificial coagulation activation must be avoided, which is relevant for the patient, for example to prevent stent graft occlusion. The aim was to evaluate the advantages and disadvantages of antithrombotic and antifouling surface coatings in the context of their suitability for ex vivo incubation and the study of coagulation properties. METHODS We investigated the impact of different protocols for surface coating of synthetic material and different anticoagulants on hemostasis and platelet activation in ex vivo human whole blood. Blood samples from healthy donors were incubated in coated microtubes on a rotating wheel at 37°C. Two protocols for surface coating were analyzed for hemostatic parameters and metabolic status, a heparin-based coating (CHC, Corline Heparin Conjugate) without further anticoagulation and a passivating coating (MPC, 2-methacryloyloxethyl phosphorylcholine) with added anticoagulants (enoxaparin, ENOX; or fondaparinux, FPX). Employing the MPC-based coating, the anticoagulants enoxaparin and fondaparinux were compared regarding their differential effects on plasmatic coagulation by thrombelastometry and on platelet activation by flowcytometry and platelet function assays. RESULTS Using the CHC coating, significant coagulation cascade activation was observed, whereas parameters remained mostly unchanged with MPC-based protocols. Extended incubation caused significantly elevated levels of the soluble membrane attack complex. Neither ENOX nor FPX caused a relevant impairment of platelet function or activation capacity and thrombelastometric parameters remained unchanged with both protocols. For translational purposes, we additionally modeled endotoxemia with the MPC-based protocols by incubating with lipopolysaccharide plus/minus thrombin. While coagulation parameters remained unchanged, elevated Interleukin 8 and Matrix Metalloproteinase 9 demonstrated preserved immune cell responsiveness. CONCLUSIONS The MPC-based protocols demonstrated better hemocompatibility compared to CHC, and ENOX and FPX proved useful for additional anticoagulation. Furthermore, this simple-to-use whole blood model may be useful for experimental analyses of the early coagulatory and immunological response without decalcification.
Collapse
Affiliation(s)
- Doreen Tabea Spiegelburg
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Marco Mannes
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Anke Schultze
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frieder Scheibenberger
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Frederik Müller
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Amadeo Klitzing
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
- Department of Transfusion Medicine and Hemostaseology, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kristina Nilsson Ekdahl
- Centre of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Bo Nilsson
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christian Karl Braun
- Institute for Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
11
|
Sloos PH, Vulliamy P, van 't Veer C, Gupta AS, Neal MD, Brohi K, Juffermans NP, Kleinveld DJB. Platelet dysfunction after trauma: From mechanisms to targeted treatment. Transfusion 2022; 62 Suppl 1:S281-S300. [PMID: 35748694 PMCID: PMC9546174 DOI: 10.1111/trf.16971] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Pieter H. Sloos
- Department of Intensive Care Medicine, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Paul Vulliamy
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Anirban Sen Gupta
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Matthew D. Neal
- Pittsburgh Trauma and Transfusion Medicine Research Center and Division of Trauma and Acute Care SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Karim Brohi
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Nicole P. Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Intensive Care MedicineOLVG HospitalAmsterdamThe Netherlands
| | - Derek J. B. Kleinveld
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Intensive Care MedicineErasmus MCRotterdamThe Netherlands
| |
Collapse
|
12
|
Aso K, Soutome T, Satoh M, Aoki T, Ogura H, Yamamoto T, Kanno H, Takahashi H. Association of autosomal-recessive-type distal renal tubular acidosis and Glanzmann thrombasthenia as a consequence of runs of homozygosity. Clin Case Rep 2022; 10:e6070. [PMID: 35865781 PMCID: PMC9295683 DOI: 10.1002/ccr3.6070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
We report the case of a Filipino girl with autosomal-recessive-type distal renal tubular acidosis and Glanzmann thrombasthenia caused by homozygous variants in the genes SLC4A1 and ITGA2B within the long homozygous DNA region on chromosome 17q21.31. This haplotype may be retained among individuals of Filipino descent.
Collapse
Affiliation(s)
- Keiko Aso
- Department of PediatricsToho University Omori Medical CenterTokyoJapan
| | - Takehiko Soutome
- Department of PediatricsToho University Omori Medical CenterTokyoJapan
| | - Mari Satoh
- Department of PediatricsToho University Omori Medical CenterTokyoJapan
| | - Takako Aoki
- Department of Transfusion Medicine and Cell ProcessingTokyo Women's Medical UniversityTokyoJapan
| | - Hiromi Ogura
- Department of Transfusion Medicine and Cell ProcessingTokyo Women's Medical UniversityTokyoJapan
| | | | - Hitoshi Kanno
- Department of Transfusion Medicine and Cell ProcessingTokyo Women's Medical UniversityTokyoJapan
| | | |
Collapse
|
13
|
Prat NJ, Meyer AD, Scaravilli V, Cannon J, Cancio LC, Cap AP, Batchinsky AI. Regional blood acidification inhibits coagulation during extracorporeal carbon dioxide removal (ECCO 2 R). Artif Organs 2022; 46:1181-1191. [PMID: 35289412 DOI: 10.1111/aor.14233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/17/2022] [Accepted: 03/04/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Consumption of platelets and coagulation factors during extracorporeal carbon dioxide removal (ECCO2 R) increases bleeding complications and associated mortality. Regional infusion of lactic acid enhances ECCO2 R by shifting the chemical equilibrium from bicarbonate to carbon dioxide. Our goal was to test if regional blood acidification during ECCO2 R inhibits platelet function and coagulation. METHODS An ECCO2 R system containing a hemofilter circulated blood at 0.25 L/min in 8 healthy ewes (Ovis aries) for 36 hours. Three of the sheep received ECCO2 R with no recirculation compared to 5 sheep that received ECCO2 R plus 12 hours of regional blood acidification via the hemofilter, placed upstream from the oxygenator, into which 4.4 M lactic acid was infused. Blood gases, platelet count and function, thromboelastography, coagulation-factor activity, and von Willebrand factor activity (vWF:Ag) were measured at baseline, at start of lactic acid infusion, and after 36 hours of extracorporeal circulation. RESULTS Twelve hours of regional acid infusion significantly inhibited platelet aggregation response to adenosine diphosphate; vWF; and platelet expression of P-selectin compared to control. It also significantly reduced consumption of fibrinogen and of coagulation factors V, VII, IX, compared to control. CONCLUSIONS Regional acidification reduces platelet activation and vitamin-K-dependent coagulation-factor consumption during ECCO2 R. This is the first report of a simple method that may enhance effective anticoagulation for ECCO2 R.
Collapse
Affiliation(s)
- Nicolas J Prat
- French Armed Forces Biomedical Research Institute (IRBA), Paris, France.,U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Andrew D Meyer
- Division of Critical Care Medicine, Department of Pediatrics, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA.,U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Vittorio Scaravilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Jeremy Cannon
- Division of Traumatology, Surgical Critical Care & Emergency Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leopoldo C Cancio
- U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Andrew P Cap
- U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | | |
Collapse
|
14
|
Pluta K, Porębska K, Urbanowicz T, Gąsecka A, Olasińska-Wiśniewska A, Targoński R, Krasińska A, Filipiak KJ, Jemielity M, Krasiński Z. Platelet-Leucocyte Aggregates as Novel Biomarkers in Cardiovascular Diseases. BIOLOGY 2022; 11:biology11020224. [PMID: 35205091 PMCID: PMC8869671 DOI: 10.3390/biology11020224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/18/2022]
Abstract
Simple Summary Cardiovascular diseases are the most common cause of death worldwide. Hence, novel biomarkers are urgently needed to improve diagnosis and treatment. Platelet–leucocyte aggregates are conglomerates of platelets and leucocytes and are widely investigated as biomarkers in cardiovascular diseases. Platelet–leucocytes aggregates are present in health, but increase in patients with cardiovascular risk factors and acute or stable coronary syndromes, making them a potential diagnostic marker. Moreover, platelet–leucocyte aggregates predict outcomes after surgery or percutaneous treatment and could be used to monitor antiplatelet therapy. Emerging data about the participation of platelet–leucocyte aggregates in cardiovascular diseases pathogenesis make them an attractive target for novel therapies. Furthermore, simple detection with conventional flow cytometry provides accurate and reproducible results, although requires specific sample handling. The main task for the future is to determine the standardized protocol to measure blood concentrations of platelet–leucocyte aggregates and subsequently establish their normal range in health and disease. Abstract Platelet–leucocyte aggregates (PLA) are a formation of leucocytes and platelets bound by specific receptors. They arise in the condition of sheer stress, thrombosis, immune reaction, vessel injury, and the activation of leukocytes or platelets. PLA participate in cardiovascular diseases (CVD). Increased levels of PLA were revealed in acute and chronic coronary syndromes, carotid stenosis cardiovascular risk factors. Due to accessible, available, replicable, quick, and low-cost quantifying using flow cytometry, PLA constitute an ideal biomarker for clinical practice. PLA are promising in early diagnosing and estimating prognosis in patients with acute or chronic coronary syndromes treated by percutaneous coronary intervention (PCI) and coronary artery bypass grafting (CABG). PLA were also a reliable marker of platelet activity for monitoring antiplatelet therapy. PLA consist also targets potential therapies in CVD. All of the above potential clinical applications require further studies to validate methods of assay and proof clinical benefits.
Collapse
Affiliation(s)
- Kinga Pluta
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (K.P.); (K.P.)
| | - Kinga Porębska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (K.P.); (K.P.)
| | - Tomasz Urbanowicz
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (M.J.)
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (K.P.); (K.P.)
- Correspondence: ; Tel.: +48-22-599-1951
| | - Anna Olasińska-Wiśniewska
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (M.J.)
| | - Radosław Targoński
- 1st Department of Cardiology, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Aleksandra Krasińska
- Department of Ophtalmology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Krzysztof J. Filipiak
- Department of Clinical Sciences, Maria Sklodowska-Curie Medical Academy in Warsaw, 00-136 Warsaw, Poland;
| | - Marek Jemielity
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (T.U.); (A.O.-W.); (M.J.)
| | - Zbigniew Krasiński
- Department of Vascular and Endovascular Surgery, Angiology and Phlebology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| |
Collapse
|
15
|
Lanier CJ, Taintor JS, Christopherson PW, Spangler EA. Effect of lactic acid addition to equine whole blood on platelet aggregation measured by impedance aggregometry. Vet Clin Pathol 2022; 51:65-69. [PMID: 34989029 DOI: 10.1111/vcp.13034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Acidemia in sick or injured horses is often due to lactic acid accumulation. Alterations in platelet function and hemostasis are among numerous deleterious effects caused by decreased physiologic pH. OBJECTIVES We aimed to evaluate the effect of hyperlactatemia and resultant acidemia on platelet aggregation in equine whole blood using impedance aggregometry. METHODS Platelet aggregation was measured using the Multiplate analyzer in whole blood from 34 healthy horses at baseline and after in vitro addition of lactic acid to adjust the pH. Platelet aggregation of each sample was quantified by the area under the curve measurement reported by the Multiplate system. The association between platelet aggregation and pH was analyzed using a linear mixed-effects model. The association of baseline platelet aggregation with hematocrits (Hcts), white blood cell (WBC) counts, and platelet counts was evaluated using Pearson's correlations. RESULTS There was a significant association between acidemia and decreased platelet aggregation. No significant correlations were detected between platelet aggregation and Hct, WBC count, or platelet count. Platelet aggregation measured in healthy horses using the Multiplate analyzer showed substantial variation between animals. CONCLUSIONS Acidemia caused by the addition of lactic acid to equine whole blood was associated with a mild though statistically significant decrease in platelet aggregation. In conjunction with other factors, this change may contribute to morbidity-related disorders of hemostasis, although its precise clinical relevance is uncertain.
Collapse
Affiliation(s)
- Christopher J Lanier
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Jennifer S Taintor
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Peter W Christopherson
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Elizabeth A Spangler
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
16
|
O’Reilly D, Murphy CA, Drew R, El-Khuffash A, Maguire PB, Ainle FN, Mc Callion N. Platelets in pediatric and neonatal sepsis: novel mediators of the inflammatory cascade. Pediatr Res 2022; 91:359-367. [PMID: 34711945 PMCID: PMC8816726 DOI: 10.1038/s41390-021-01715-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
Sepsis, a dysregulated host response to infection, has been difficult to accurately define in children. Despite a higher incidence, especially in neonates, a non-specific clinical presentation alongside a lack of verified biomarkers has prevented a common understanding of this condition. Platelets, traditionally regarded as mediators of haemostasis and thrombosis, are increasingly associated with functions in the immune system with involvement across the spectrum of innate and adaptive immunity. The large number of circulating platelets (approx. 150,000 cells per microlitre) mean they outnumber traditional immune cells and are often the first to encounter a pathogen at a site of injury. There are also well-described physiological differences between platelets in children and adults. The purpose of this review is to place into context the platelet and its role in immunology and examine the evidence where available for its role as an immune cell in childhood sepsis. It will examine how the platelet interacts with both humoral and cellular components of the immune system and finally discuss the role the platelet proteome, releasate and extracellular vesicles may play in childhood sepsis. This review also examines how platelet transfusions may interfere with the complex relationships between immune cells in infection. IMPACT: Platelets are increasingly being recognised as important "first responders" to immune threats. Differences in adult and paediatric platelets may contribute to differing immune response to infections. Adult platelet transfusions may affect infant immune responses to inflammatory/infectious stimuli.
Collapse
Affiliation(s)
- Daniel O’Reilly
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
| | - Claire A. Murphy
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| | - Richard Drew
- grid.416068.d0000 0004 0617 7587Clinical Innovation Unit, Rotunda Hospital, Dublin, Ireland ,Irish Meningitis and Sepsis Reference Laboratory, Children’s Health Ireland at Temple Street, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Afif El-Khuffash
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| | - Patricia B. Maguire
- grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland
| | - Fionnuala Ni Ainle
- grid.7886.10000 0001 0768 2743Conway-SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Biomolecular & Biomedical Science, University College Dublin, Dublin, Ireland ,grid.411596.e0000 0004 0488 8430Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland ,grid.416068.d0000 0004 0617 7587Department of Haematology, Rotunda Hospital, Dublin, Ireland ,grid.7886.10000 0001 0768 2743School of Medicine, University College Dublin, Dublin, Ireland
| | - Naomi Mc Callion
- grid.416068.d0000 0004 0617 7587Department of Neonatology, Rotunda Hospital, Dublin, Ireland ,grid.4912.e0000 0004 0488 7120Department of Paediatrics, Royal College of Surgeons in Ireland, Dubin, Ireland
| |
Collapse
|
17
|
Bhan A, Ansari K, Chen MY, Jandial R. Human induced pluripotent stem cell-derived platelets loaded with lapatinib effectively target HER2+ breast cancer metastasis to the brain. Sci Rep 2021; 11:16866. [PMID: 34654856 PMCID: PMC8521584 DOI: 10.1038/s41598-021-96351-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 08/06/2021] [Indexed: 01/09/2023] Open
Abstract
Prognosis of patients with HER2+ breast-to-brain-metastasis (BBM) is dismal even after current standard-of-care treatments, including surgical resection, whole-brain radiation, and systemic chemotherapy. Radiation and systemic chemotherapies can also induce cytotoxicity, leading to significant side effects. Studies indicate that donor-derived platelets can serve as immune-compatible drug carriers that interact with and deliver drugs to cancer cells with fewer side effects, making them a promising therapeutic option with enhanced antitumor activity. Moreover, human induced pluripotent stem cells (hiPSCs) provide a potentially renewable source of clinical-grade transfusable platelets that can be drug-loaded to complement the supply of donor-derived platelets. Here, we describe methods for ex vivo generation of megakaryocytes (MKs) and functional platelets from hiPSCs (hiPSC-platelets) in a scalable fashion. We then loaded hiPSC-platelets with lapatinib and infused them into BBM tumor-bearing NOD/SCID mouse models. Such treatment significantly increased intracellular lapatinib accumulation in BBMs in vivo, potentially via tumor cell-induced activation/aggregation. Lapatinib-loaded hiPSC-platelets exhibited normal morphology and function and released lapatinib pH-dependently. Importantly, lapatinib delivery to BBM cells via hiPSC-platelets inhibited tumor growth and prolonged survival of tumor-bearing mice. Overall, use of lapatinib-loaded hiPSC-platelets effectively reduced adverse effects of free lapatinib and enhanced its therapeutic efficacy, suggesting that they represent a novel means to deliver chemotherapeutic drugs as treatment for BBM.
Collapse
Affiliation(s)
- Arunoday Bhan
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| | - Khairul Ansari
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
- Celcuity LLC, Minneapolis, MN, 55446, USA
| | - Mike Y Chen
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope Medical Center, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| |
Collapse
|
18
|
Jansen EE, Hartmann M. Clot Retraction: Cellular Mechanisms and Inhibitors, Measuring Methods, and Clinical Implications. Biomedicines 2021; 9:1064. [PMID: 34440268 PMCID: PMC8394358 DOI: 10.3390/biomedicines9081064] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 11/22/2022] Open
Abstract
Platelets have important functions in hemostasis. Best investigated is the aggregation of platelets for primary hemostasis and their role as the surface for coagulation leading to fibrin- and clot-formation. Importantly, the function of platelets does not end with clot formation. Instead, platelets are responsible for clot retraction through the concerted action of the activated αIIbβ3 receptors on the surface of filopodia and the platelet's contractile apparatus binding and pulling at the fibrin strands. Meanwhile, the signal transduction events leading to clot retraction have been investigated thoroughly, and several targets to inhibit clot retraction have been demonstrated. Clot retraction is a physiologically important mechanism allowing: (1) the close contact of platelets in primary hemostasis, easing platelet aggregation and intercellular communication, (2) the reduction of wound size, (3) the compaction of red blood cells to a polyhedrocyte infection-barrier, and (4) reperfusion in case of thrombosis. Several methods have been developed to measure clot retraction that have been based on either the measurement of clot volume or platelet forces. Concerning the importance of clot retraction in inborn diseases, the failure of clot retraction in Glanzmann thrombasthenia is characterized by a bleeding phenotype. Concerning acquired diseases, altered clot retraction has been demonstrated in patients with coronary heart disease, stroke, bronchial asthma, uremia, lupus erythematodes, and other diseases. However, more studies on the diagnostic and prognostic value of clot retraction with methods that have to be standardized are necessary.
Collapse
Affiliation(s)
- Ellen E. Jansen
- Clinic for Operative Dentistry, Periodontology and Preventive Dentistry, RWTH Aachen University, 52074 Aachen, Germany;
| | - Matthias Hartmann
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
19
|
Johnson L, Vekariya S, Wood B, Tan S, Roan C, Marks DC. Refrigeration of apheresis platelets in platelet additive solution (PAS-E) supports in vitro platelet quality to maximize the shelf-life. Transfusion 2021; 61 Suppl 1:S58-S67. [PMID: 34269458 DOI: 10.1111/trf.16489] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Refrigeration, or cold-storage, of platelets may be beneficial to extend the limited shelf-life of conventionally stored platelets and support transfusion protocols in rural and military areas. The aim of this study was to compare the morphologic, metabolic, and functional aspects of apheresis platelets stored at room-temperature (RT) or cold conditions, in either plasma or supplemented with platelet additive solution (PAS). STUDY DESIGN AND METHODS Double-dose apheresis platelets were collected in either 100% plasma or 40% plasma/60% PAS-E using the Trima apheresis platform. One component from each group was either stored at RT (20-24°C) or refrigerated (2-6°C). Platelets were tested over a 21-day period. RESULTS The platelet concentration decreased by approximately 30% in all groups during 21 days of storage (p > .05). Cold-storage reduced glycolytic metabolism, and the pH was maintained above the minimum specification (>6.4) for 21 days only when platelets were stored in PAS. The surface phenotype and the composition of the supernatant were differentially affected by temperature and storage solution. Functional responses (aggregation, agonist-induced receptor activation, clotting time) were improved during cold-storage, and the influence of residual plasma was assay dependent. CONCLUSION In vitro platelet quality is differentially affected by storage time, temperature, and solution. Cold-storage, particularly in PAS, better maintains key metabolic, phenotypic, and functional parameters during prolonged storage.
Collapse
Affiliation(s)
- Lacey Johnson
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| | - Shuchna Vekariya
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| | - Ben Wood
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| | - Shereen Tan
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| | - Christopher Roan
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| | - Denese C Marks
- Research and Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
20
|
Ghirardello S, Lecchi A, Artoni A, Panigada M, Aliberti S, Scalambrino E, La Marca S, Boscarino M, Gramegna A, Properzi P, Abruzzese C, Blasi F, Grasselli G, Mosca F, Tripodi A, Peyvandi F. Assessment of Platelet Thrombus Formation under Flow Conditions in Adult Patients with COVID-19: An Observational Study. Thromb Haemost 2021; 121:1087-1096. [PMID: 33545735 DOI: 10.1055/s-0041-1722919] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with systemic inflammation, which may dysregulate platelet function. Total Thrombus-Formation Analysis System (T-TAS) is a flow-chamber device that analyses platelet-mediated thrombus formation in capillary channels through the following parameters: (1) the area under the flow-pressure curve (AUC), (2) occlusion start time (OST), time needed to reach OST, and (3) occlusion time (OT), time needed to reach the occlusion pressure. METHODS AND FINDINGS Sixty-one COVID-19 patients admitted to intensive, subintensive, and low intensive care were prospectively enrolled according to the time of admission: group A (up to 8 days) (n = 18); group B (from 9 to 21 days) (n = 19), and group C ( > 21 days) (n = 24). T-TAS measurements were performed at enrolment and after 7 days. Median OST was similar among groups. AUC was lower in group A compared to B (p = 0.001) and C (p = 0.033). OT was longer in group A compared to B (p = 0.001) and C (p = 0.028). Platelet count (PC) was higher in group B compared to A (p = 0.024). The linear regression showed that OT and AUC were independent from PC in group A (OT: 0.149 [95% confidence interval [CI]: -0.326 to 0.624], p = 0.513 and AUC: 0.005 [95% CI: -0.008 to 0.017], p = 0,447). In contrast, in group B, PC was associated with OT (-0.019 [-0.028 to 0.008], p = 0.023) and AUC (0.749 [0.358-1.139], p = 0,015), similarly to group C. Conversely, patients with different illness severity had similar T-TAS parameters. CONCLUSION COVID-19 patients display an impaired platelet thrombus formation in the early phase of the disease compared to later stages and controls, independently from illness severity.
Collapse
Affiliation(s)
- Stefano Ghirardello
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Lecchi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Artoni
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Panigada
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore, Milan, Italy
| | - Stefano Aliberti
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Erica Scalambrino
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia La Marca
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Boscarino
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Gramegna
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Properzi
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore, Milan, Italy
| | - Chiara Abruzzese
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore, Milan, Italy
| | - Francesco Blasi
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Grasselli
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Armando Tripodi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Flora Peyvandi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
21
|
Lawson CA, Spangler EA. The effect of adding lactic acid to canine whole blood on platelet aggregation as measured by impedance aggregometry. Vet Clin Pathol 2020; 49:217-221. [PMID: 32535936 DOI: 10.1111/vcp.12862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/22/2019] [Accepted: 10/25/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Acidemia in sick dogs often results from the accumulation of lactic acid. The resulting decrease in blood pH can have many physiologic effects, including alteration of platelet function. OBJECTIVES We aimed to evaluate the effect of hyperlactatemia and subsequent acidemia on platelet aggregation in canine blood using impedance aggregometry. METHODS Platelet aggregation was measured in blood from 27 healthy dogs using the Multiplate analyzer at baseline and after in vitro addition of two different volumes of lactic acid to adjust the pH. The area under the curve (AUC), reported by the Multiplate analyzer, was used to assess the extent of platelet aggregation in each sample. A linear mixed effects model was used to test for the association between platelet aggregation and pH. The association of baseline platelet aggregation with HCTs, platelet counts, and WBC counts was assessed using Pearson's correlations. RESULTS Acidemia was associated with a significant decrease in platelet aggregation. No significant correlations were detected between platelet aggregation and HCT, platelet count, or WBC count. Platelet aggregation measured using the Multiplate analyzer showed substantial individual variation. CONCLUSIONS Worsening acidemia due to the addition of lactic acid caused a mild but significant decrease in platelet aggregation in canine blood. The clinical significance of this change is uncertain but could be important when combined with other abnormalities of hemostasis associated with illness.
Collapse
Affiliation(s)
- Cheryl A Lawson
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Elizabeth A Spangler
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| |
Collapse
|
22
|
Wu YW, Huang CC, Changou CA, Lu LS, Goubran H, Burnouf T. Clinical-grade cryopreserved doxorubicin-loaded platelets: role of cancer cells and platelet extracellular vesicles activation loop. J Biomed Sci 2020; 27:45. [PMID: 32200762 PMCID: PMC7087392 DOI: 10.1186/s12929-020-00633-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Human platelets (PLT) and PLT-extracellular vesicles (PEV) released upon thrombin activation express receptors that interact with tumour cells and, thus, can serve as a delivery platform of anti-cancer agents. Drug-loaded nanoparticles coated with PLT membranes were demonstrated to have improved targeting efficiency to tumours, but remain impractical for clinical translation. PLT and PEV targeted drug delivery vehicles should facilitate clinical developments if clinical-grade procedures can be developed. METHODS PLT from therapeutic-grade PLT concentrate (PC; N > 50) were loaded with doxorubicin (DOX) and stored at - 80 °C (DOX-loaded PLT) with 6% dimethyl sulfoxide (cryopreserved DOX-loaded PLT). Surface markers and function of cryopreserved DOX-loaded PLT was confirmed by Western blot and thromboelastography, respectively. The morphology of fresh and cryopreserved naïve and DOX-loaded PLT was observed by scanning electron microscopy. The content of tissue factor-expressing cancer-derived extracellular vesicles (TF-EV) present in conditioned medium (CM) of breast cancer cells cultures was measured. The drug release by fresh and cryopreserved DOX-loaded PLT triggered by various pH and CM was determined by high performance liquid chromatography. The thrombin activated PEV was analyzed by nanoparticle tracking analysis. The cellular uptake of DOX from PLT was observed by deconvolution microscopy. The cytotoxicities of DOX-loaded PLT, cryopreserved DOX-loaded PLT, DOX and liposomal DOX on breast, lung and colon cancer cells were analyzed by CCK-8 assay. RESULTS 15~36 × 106 molecules of DOX could be loaded in each PLT within 3 to 9 days after collection. The characterization and bioreactivity of cryopreserved DOX-loaded PLT were preserved, as evidenced by (a) microscopic observations, (b) preservation of important PLT membrane markers CD41, CD61, protease activated receptor-1, (c) functional activity, (d) reactivity to TF-EV, and (e) efficient generation of PEV upon thrombin activation. The transfer of DOX from cryopreserved PLT to cancer cells was achieved within 90 min, and stimulated by TF-EV and low pH. The cryopreserved DOX-loaded PLT formulation was 7~23-times more toxic to three cancer cells than liposomal DOX. CONCLUSIONS Cryopreserved DOX-loaded PLT can be prepared under clinically compliant conditions preserving the membrane functionality for anti-cancer therapy. These findings open perspectives for translational applications of PLT-based drug delivery systems.
Collapse
Affiliation(s)
- Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
| | - Cheng-Chain Huang
- Graduate Institute of Translational Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun Austin Changou
- Graduate Institute of Translational Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
23
|
Oneto P, Zubiry PR, Schattner M, Etulain J. Anticoagulants Interfere With the Angiogenic and Regenerative Responses Mediated by Platelets. Front Bioeng Biotechnol 2020; 8:223. [PMID: 32266247 PMCID: PMC7098916 DOI: 10.3389/fbioe.2020.00223] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/05/2020] [Indexed: 12/29/2022] Open
Abstract
Background and aims Platelet rich plasma (PRP) obtained from blood anticoagulated with acid-citrate-dextrose (ACD) or sodium-citrate (SC) is used for regenerative medicine as source of platelet-derived growth factors. Allergic reactions against citrate were reported in patients after local injection of PRP allowing us to hypothesize that anticoagulants exert a harmful and local effect that interferes with the regenerative proprieties of platelets. Herein we test this hypothesis by analyzing the effect of ACD and SC on angiogenic and regenerative responses mediated by platelets. Methods PRP was obtained from SC- or ACD-anticoagulated blood; platelets were lysed to release growth factors; and PRP releasates (PRPr) were used to induce in vitro endothelial proliferation and 2D-migration, and regeneration of mouse skin wounds. Results We first compared proliferation and migration of endothelial cells mediated by anticoagulated-PRPr supplemented or not with CaCl2. Alteration of endothelial adhesion and impediment of proliferation and migration was observed without CaCl2. Although endothelial morphology was normalized in SC- and ACD-PRPr after calcium restitution, angiogenic responses were only markedly induced by SC-PRPr. In vivo studies revealed a delay in mouse skin regeneration after treatment with anticoagulated-PRPr without CaCl2. Healing was only induced after calcium restitution in SC- but ACD-PRPr. Moreover, the development of inflammatory intradermal papules was evidenced after injection of ACD-PRPr. Supplementation of SC-PRPr with the equivalent concentration of dextrose (D-Glucose, 18 mM) present in ACD-PRPr resulted in reduction of endothelial proliferation and migration, delay of mouse skin regeneration and development of intradermal papules. Finally, collecting blood with half amount of SC significantly improved all the angiogenic and regenerative responses mediated by PRPr. In contrast, the delay of skin regeneration and the development of inflammatory papules remained stable after dilution of ACD. Conclusion Our findings indicate that (1) calcium restitution is required to impair the cellular and tissue alterations induced by citrated-anticoagulants contained in PRP; (2) ACD-derived dextrose confers anti-angiogenic, anti-regenerative and pro-inflammatory proprieties to PRP; and (3) half concentration of SC improves the angiogenesis and regeneration mediated by PRP.
Collapse
Affiliation(s)
- Paula Oneto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Paula Romina Zubiry
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Julia Etulain
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
24
|
Lopez E, Srivastava AK, Burchfield J, Wang YW, Cardenas JC, Togarrati PP, Miyazawa B, Gonzalez E, Holcomb JB, Pati S, Wade CE. Platelet-derived- Extracellular Vesicles Promote Hemostasis and Prevent the Development of Hemorrhagic Shock. Sci Rep 2019; 9:17676. [PMID: 31776369 PMCID: PMC6881357 DOI: 10.1038/s41598-019-53724-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 11/04/2019] [Indexed: 01/01/2023] Open
Abstract
Every year more than 500,000 deaths are attributed to trauma worldwide and severe hemorrhage is present in most of them. Transfused platelets have been shown to improve survival in trauma patients, although its mechanism is only partially known. Platelet derived-extracellular vesicles (PEVs) are small vesicles released from platelets upon activation and/or mechanical stimulation and many of the benefits attributed to platelets could be mediated through PEVs. Based on the available literature, we hypothesized that transfusion of human PEVs would promote hemostasis, reduce blood loss and attenuate the progression to hemorrhagic shock following severe trauma. In this study, platelet units from four different donors were centrifuged to separate platelets and PEVs. The pellets were washed to obtain plasma-free platelets to use in the rodent model. The supernatant was subjected to tangential flow filtration for isolation and purification of PEVs. PEVs were assessed by total count and particle size distribution by Nanoparticle Tracking Analysis (NTA) and characterized for cells of origin and expression of EV specific-surface and cytosolic markers by flow cytometry. The coagulation profile from PEVs was assessed by calibrated automated thrombography (CAT) and thromboelastography (TEG). A rat model of uncontrolled hemorrhage was used to compare the therapeutic effects of 8.7 × 108 fresh platelets (FPLT group, n = 8), 7.8 × 109 PEVs (PEV group, n = 8) or Vehicle (Control, n = 16) following severe trauma. The obtained pool of PEVs from 4 donors had a mean size of 101 ± 47 nm and expressed the platelet-specific surface marker CD41 and the EV specific markers CD9, CD61, CD63, CD81 and HSP90. All PEV isolates demonstrated a dose-dependent increase in the rate and amount of thrombin generated and overall clot strength. In vivo experiments demonstrated a 24% reduction in abdominal blood loss following liver trauma in the PEVs group when compared with the control group (9.9 ± 0.4 vs. 7.5 ± 0.5 mL, p < 0.001>). The PEV group also exhibited improved outcomes in blood pressure, lactate level, base excess and plasma protein concentration compared to the Control group. Fresh platelets failed to improve these endpoints when compared to Controls. Altogether, these results indicate that human PEVs provide pro-hemostatic support following uncontrolled bleeding. As an additional therapeutic effect, PEVs improve the outcome following severe trauma by maintaining hemodynamic stability and attenuating the development of ischemia, base deficit, and cardiovascular shock.
Collapse
Affiliation(s)
- Ernesto Lopez
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA.
| | - Amit K Srivastava
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - John Burchfield
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Yao-Wei Wang
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Jessica C Cardenas
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | | | - Byron Miyazawa
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Erika Gonzalez
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - John B Holcomb
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| | - Shibani Pati
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Charles E Wade
- Center for Translational Injury Research (CeTIR), Department of Surgery, University of Texas Health Science Center at Houston, Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
25
|
Platelets in Host Defense: Experimental and Clinical Insights. Trends Immunol 2019; 40:922-938. [PMID: 31601520 DOI: 10.1016/j.it.2019.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022]
Abstract
Platelets are central players in thrombosis and hemostasis but are increasingly recognized as key components of the immune system. They shape ensuing immune responses by recruiting leukocytes, and support the development of adaptive immunity. Recent data shed new light on the complex role of platelets in immunity. Here, we summarize experimental and clinical data on the role of platelets in host defense against bacteria. Platelets bind, contain, and kill bacteria directly; however, platelet proinflammatory effector functions and cross-talk with the coagulation system, can also result in damage to the host (e.g., acute lung injury and sepsis). Novel clinical insights support this dichotomy: platelet inhibition/thrombocytopenia can be either harmful or protective, depending on pathophysiological context. Clinical studies are currently addressing this aspect in greater depth.
Collapse
|
26
|
Esiaba I, Mousselli I, M. Faison G, M. Angeles D, S. Boskovic D. Platelets in the Newborn. NEONATAL MEDICINE 2019. [DOI: 10.5772/intechopen.86715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
27
|
|
28
|
Karakatsanis SJ, Papadatos SS, Syrigos KN. Hypoplastic thrombocytopenia and platelet transfusion: therapeutic goals. Hosp Pract (1995) 2019; 47:16-23. [PMID: 30409035 DOI: 10.1080/21548331.2019.1546530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/07/2018] [Indexed: 06/08/2023]
Abstract
Platelet transfusions consist a major part of the management of hypoplastic thrombocytopenia, the latter occurring mainly among patients with hematological malignancies. Platelet transfusions have led to a reduction of deaths attributable to thrombocytopenia-induced bleeding, despite their possible complications; nonetheless, prophylactic administration of platelets to patients with severe thrombocytopenia or before invasive procedures should be based on specific criteria, as well as therapeutic administration during active bleeding. Recently developed ex-vivo procedures have resulted in producing safer blood products, yet it remains unclear whether these pathogen-inactivated products have sufficient efficacy. What is more, another significant problem that remains to be more effectively addressed is the developing refractoriness to platelet transfusions.
Collapse
Affiliation(s)
- Stamatis J Karakatsanis
- a Faculty of Medicine, Hematology Unit, 3rd Department of Internal Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Stamatis S Papadatos
- b Faculty of Medicine, Internal Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Konstantinos N Syrigos
- c Faculty of Medicine, 3rd Department of Internal Medicine , National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
29
|
Unravelling the Interplay between Extracellular Acidosis and Immune Cells. Mediators Inflamm 2018; 2018:1218297. [PMID: 30692870 PMCID: PMC6332927 DOI: 10.1155/2018/1218297] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 11/28/2018] [Indexed: 01/18/2023] Open
Abstract
The development of an acidic tissue environment is a hallmark of a variety of inflammatory processes and solid tumors. However, little attention has been paid so far to analyze the influence exerted by extracellular pH on the immune response. Tissue acidosis (pH 6.0 to 7.0) is usually associated with the course of infectious processes in peripheral tissues. Moreover, it represents a prominent feature of solid tumors. In fact, values of pH ranging from 5.7 to 7.0 are usually found in a number of solid tumors such as breast cancer, brain tumors, sarcomas, malignant melanoma, squamous cell carcinomas, and adenocarcinomas. Both the innate and adaptive arms of the immune response appear to be finely regulated by extracellular acidosis in the range of pH values found at inflammatory sites and tumors. Low pH has been shown to delay neutrophil apoptosis, promoting their differentiation into a proangiogenic profile. Acting on monocytes and macrophages, it induces the activation of the inflammasome and the production of IL-1β, while the exposure of conventional dendritic cells to low pH promotes the acquisition of a mature phenotype. Overall, these observations suggest that high concentrations of protons could be recognized by innate immune cells as a danger-associated molecular pattern (DAMP). On the other hand, by acting on T lymphocytes, low pH has been shown to suppress the cytotoxic response mediated by CD8+ T cells as well as the production of IFN-γ by TH1 cells. Interestingly, modulation of tumor microenvironment acidity has been shown to be able not only to reverse anergy in human and mouse tumor-infiltrating T lymphocytes but also to improve the antitumor immune response induced by checkpoint inhibitors. Here, we provide an integrated view of the influence exerted by low pH on immune cells and discuss its implications in the immune response against infectious agents and tumor cells.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW This review describes the essential roles of platelets in neutrophil recruitment from the bloodstream into inflamed and infected tissues, with a focus on recent findings. RECENT FINDINGS Platelets are required for the recruitment of neutrophils to sites of inflammation and infection. They fulfil this role largely by enabling contacts of circulating neutrophils with the inflamed blood vessel wall prior to extravasation. Platelets promote both early stages of neutrophil recruitment (tethering, rolling, arrest, firm adhesion) and - as recent work has demonstrated - later stages (intravascular crawling and diapedesis). Recent studies have also begun to identify platelet-signaling pathways that can elicit the underlying interactions between platelets, neutrophils and vascular endothelial cells without stimulating concomitant platelet aggregation and thrombus formation. These pathways include Rho-guanine-nucleotide binding proteins and Rho-guanine-nucleotide exchange factors. SUMMARY Recent findings have contributed to our burgeoning understanding of the platelet-dependent mechanisms that control neutrophil recruitment to sites of inflammation and have opened up new avenues of research aimed at increasing our knowledge of these mechanisms further. These insights might lead to the development of novel anti-inflammatory drugs that will be useful in a wide range of inflammatory diseases without causing immunodeficiency.
Collapse
|
31
|
Abrey Recalde MJ, Alvarez RS, Alberto F, Mejias MP, Ramos MV, Fernandez Brando RJ, Bruballa AC, Exeni RA, Alconcher L, Ibarra CA, Amaral MM, Palermo MS. Soluble CD40 Ligand and Oxidative Response Are Reciprocally Stimulated during Shiga Toxin-Associated Hemolytic Uremic Syndrome. Toxins (Basel) 2017; 9:toxins9110331. [PMID: 29068360 PMCID: PMC5705951 DOI: 10.3390/toxins9110331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/29/2017] [Accepted: 10/15/2017] [Indexed: 01/01/2023] Open
Abstract
Shiga toxin (Stx), produced by Escherichia coli, is the main pathogenic factor of diarrhea-associated hemolytic uremic syndrome (HUS), which is characterized by the obstruction of renal microvasculature by platelet-fibrin thrombi. It is well known that the oxidative imbalance generated by Stx induces platelet activation, contributing to thrombus formation. Moreover, activated platelets release soluble CD40 ligand (sCD40L), which in turn contributes to oxidative imbalance, triggering the release of reactive oxidative species (ROS) on various cellular types. The aim of this work was to determine if the interaction between the oxidative response and platelet-derived sCD40L, as consequence of Stx-induced endothelium damage, participates in the pathogenic mechanism during HUS. Activated human glomerular endothelial cells (HGEC) by Stx2 induced platelets to adhere to them. Although platelet adhesion did not contribute to endothelial damage, high levels of sCD40L were released to the medium. The release of sCD40L by activated platelets was inhibited by antioxidant treatment. Furthermore, we found increased levels of sCD40L in plasma from HUS patients, which were also able to trigger the respiratory burst in monocytes in a sCD40L-dependent manner. Thus, we concluded that platelet-derived sCD40L and the oxidative response are reciprocally stimulated during Stx2-associated HUS. This process may contribute to the evolution of glomerular occlusion and the microangiopathic lesions.
Collapse
Affiliation(s)
- Maria J Abrey Recalde
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Romina S Alvarez
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica "Bernardo Houssay", Facultad de Medicina-Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, 1121 Buenos Aires, Argentina.
| | - Fabiana Alberto
- División Trombosis, Instituto de investigaciones Hematológicas "Mariano R. Castex", Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Maria P Mejias
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Maria V Ramos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Romina J Fernandez Brando
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Andrea C Bruballa
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| | - Ramon A Exeni
- Departamento de Nefrología, Hospital Municipal del Niño, San Justo, B1754FUD Provincia de Buenos Aires, Argentina.
| | - Laura Alconcher
- Unidad de Nefrourología Infantil. Hospital Interzonal General Dr. José Penna, Bahía Blanca, 8000 Provincia de Buenos Aires, Argentina.
| | - Cristina A Ibarra
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica "Bernardo Houssay", Facultad de Medicina-Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, 1121 Buenos Aires, Argentina.
| | - María M Amaral
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Instituto de Fisiología y Biofísica "Bernardo Houssay", Facultad de Medicina-Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, 1121 Buenos Aires, Argentina.
| | - Marina S Palermo
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas-Academia Nacional de Medicina, 1425 Buenos Aires, Argentina.
| |
Collapse
|
32
|
Fisher R, Lei K, Mitchell MJ, Moore GW, Dickie H, Tovey L, Crichton S, Ostermann M. The effect of regional citrate anti-coagulation on the coagulation system in critically ill patients receiving continuous renal replacement therapy for acute kidney injury - an observational cohort study. BMC Nephrol 2017; 18:304. [PMID: 28969597 PMCID: PMC5625709 DOI: 10.1186/s12882-017-0718-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/19/2017] [Indexed: 11/25/2022] Open
Abstract
Background Regional anticoagulation with citrate is the recommended first line treatment for patients receiving continuous renal replacement therapy (CRRT). There is wide variability in filter patency which may be due to differences in patient characteristics and local practice. It is also possible that citrate has effects on primary and secondary haemostasis, fibrinolysis and platelet function that are still unknown. The primary aim of the study is to describe the effect of citrate on coagulation and fibrinolysis pathways in both the patient and the haemodialysis circuit. Methods The study will recruit 12 adult patients admitted to the intensive care unit, requiring CRRT with regional citrate anticoagulation for acute kidney injury. Patients with pre-existing thrombotic or bleeding tendencies will be excluded. Thrombin generation, clot lysis and platelet function will be measured at baseline and at 12, 24, 36, 48 and 72 h after commencing CRRT (from the patient and from the circuit). We will describe the evolution of parameters over time as well as the differences in parameters between the patient and the circuit. Discussion The study will provide new data on the effects of citrate during continuous renal replacement therapy which is not currently available. We will minimise confounding factors through the use of tight exclusion criteria and accept that this will slow down recruitment. Depending on the results, we hope to incorporate the findings into existing clinical guidelines and clinical practice with the aim to prevent premature filter clotting and interruptions in treatment. Trial registration The study was registered with clinicaltrials.gov on 10th June 2015 (NCT02486614).
Collapse
Affiliation(s)
- Richard Fisher
- Guy's & St Thomas' NHS Foundation Trust, Department of Critical Care, London, SE1 9RT, UK
| | - Katie Lei
- Guy's & St Thomas' NHS Foundation Trust, Department of Critical Care, London, SE1 9RT, UK
| | - Mike J Mitchell
- St Thomas' Hospital, Molecular Haemostasis & Thrombosis, London, SE1 9RT, UK
| | - Gary W Moore
- St Thomas' Hospital, Diagnostic Haemostasis & Thrombosis Laboratory, London, SE1 9RT, UK
| | - Helen Dickie
- Guy's & St Thomas' NHS Foundation Trust, Department of Critical Care, London, SE1 9RT, UK
| | - Linda Tovey
- Guy's & St Thomas' NHS Foundation Trust, Department of Critical Care, London, SE1 9RT, UK
| | - Siobhan Crichton
- MRC Clinical Trials Unit, University College London, London, WC2B 6NH, UK
| | - Marlies Ostermann
- King's College London, Guy's & St Thomas' NHS Foundation Trust, Department of Critical Care, London, SE1 9RT, UK.
| |
Collapse
|
33
|
Potential role of platelet-leukocyte aggregation in trauma-induced coagulopathy: Ex vivo findings. J Trauma Acute Care Surg 2017; 82:921-926. [PMID: 28257393 DOI: 10.1097/ta.0000000000001410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Platelet dysfunction has been identified as an important contributor of trauma-induced coagulopathy, but the underlying mechanism still remains to be elucidated. Trauma-associated proinflammatory stimuli strongly activate leukocytes, which in turn bind activated platelets. Therefore, we investigated the role of platelet-leukocyte aggregation (PLA) as a potential feature of trauma-induced platelet dysfunction. METHODS Whole blood from 10 healthy donors was exposed to selective and collective platelet and leukocyte agonists in order to simulate differential states of activation. PLA formation and CD11b expression as a measure of leukocyte activation were determined by flow cytometry. Platelet-mediated hemostatic function was measured by thromboelastometry (ROTEM) and impedance aggregometry (Multiplate). RESULTS Activation of platelets and leukocytes was associated with diminished platelet-mediated hemostatic potential. Aggregation of platelets with monocytes rather than granulocytes resulted in a reduction of hemostatic function, as indicated by an impaired responsiveness in platelet aggregometry and a reduction of thromboelastometric maximum clot firmness. This finding was irrespective of CD11b expression and was not paralleled by a reduction of measurable platelet counts. CONCLUSION PLA formation occurs primarily between monocytes and activated platelets and is associated with impaired platelet-mediated hemostatic function. PLA formation was not paralleled by a reduction in platelet complete blood counts.
Collapse
|
34
|
Marin Oyarzún CP, Carestia A, Lev PR, Glembotsky AC, Castro Ríos MA, Moiraghi B, Molinas FC, Marta RF, Schattner M, Heller PG. Neutrophil extracellular trap formation and circulating nucleosomes in patients with chronic myeloproliferative neoplasms. Sci Rep 2016; 6:38738. [PMID: 27958278 PMCID: PMC5153854 DOI: 10.1038/srep38738] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022] Open
Abstract
The mechanisms underlying increased thrombotic risk in chronic myeloproliferative neoplasms (MPN) are incompletely understood. We assessed whether neutrophil extracellular traps (NETs), which promote thrombosis, contribute to the procoagulant state in essential thrombocythemia, polycythemia vera and myelofibrosis (MF) patients. Although MPN neutrophils showed increased basal reactive oxygen species (ROS), enhanced NETosis by unstimulated neutrophils was an infrequent finding, whereas PMA-triggered NETosis was impaired, particularly in MF, due to decreased PMA-triggered ROS production. Elevated circulating nucleosomes were a prominent finding and were higher in patients with advanced disease, which may have potential prognostic implication. Histone-MPO complexes, proposed as specific NET biomarker, were seldomly detected, suggesting NETs may not be the main source of nucleosomes in most patients, whereas their correlation with high LDH points to increased cell turn-over as a plausible origin. Lack of association of nucleosomes or NETs with thrombosis or activation markers does not support their use as predictors of thrombosis although prospective studies in a larger cohort may help define their potential contribution to MPN thrombosis. These results do not provide evidence for relevant in vivo NETosis in MPN patients under steady state conditions, although availability of standardized NET biomarkers may contribute to further research in this field.
Collapse
Affiliation(s)
- Cecilia P Marin Oyarzún
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Agostina Carestia
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX)- CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Paola R Lev
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Ana C Glembotsky
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | | | - Beatriz Moiraghi
- Department of Hematology, Hospital Ramos Mejía, Buenos Aires, Argentina
| | - Felisa C Molinas
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Rosana F Marta
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX)- CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Paula G Heller
- Department of Hematology Research, Institute of Medical Research "Alfredo Lanari", University of Buenos Aires, National Council for Scientific and Technological Research (CONICET), Buenos Aires, Argentina
| |
Collapse
|
35
|
Kareva I, Abou-Slaybi A, Dodd O, Dashevsky O, Klement GL. Normal Wound Healing and Tumor Angiogenesis as a Game of Competitive Inhibition. PLoS One 2016; 11:e0166655. [PMID: 27935954 PMCID: PMC5147849 DOI: 10.1371/journal.pone.0166655] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/01/2016] [Indexed: 12/20/2022] Open
Abstract
Both normal wound healing and tumor angiogenesis are mitigated by the sequential, carefully orchestrated release of growth stimulators and inhibitors. These regulators are released from platelet clots formed at the sites of activated endothelium in a temporally and spatially controlled manner, and the order of their release depends on their affinity to glycosaminoglycans (GAG) such as heparan sulfate (HS) within the extracellular matrix, and platelet open canallicular system. The formation of vessel sprouts, triggered by angiogenesis regulating factors with lowest affinities for heparan sulfate (e.g. VEGF), is followed by vessel-stabilizing PDGF-B or bFGF with medium affinity for HS, and by inhibitors such as PF-4 and TSP-1 with the highest affinities for HS. The invasive wound-like edge of growing tumors has an overabundance of angiogenesis stimulators, and we propose that their abundance out-competes angiogenesis inhibitors, effectively preventing inhibition of angiogenesis and vessel maturation. We evaluate this hypothesis using an experimentally motivated agent-based model, and propose a general theoretical framework for understanding mechanistic similarities and differences between the processes of normal wound healing and pathological angiogenesis from the point of view of competitive inhibition.
Collapse
Affiliation(s)
- Irina Kareva
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Mathematical, Computational and Modeling Sciences Center, Arizona State Univ, Tempe, Arizona, United States of America
| | - Abdo Abou-Slaybi
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Oliver Dodd
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Massachusetts Institute of Technology, Boston, Massachusetts, United States of America
| | - Olga Dashevsky
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Dept. of Medical Oncology, Dana−Farber Cancer Institute, Dept. of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Giannoula Lakka Klement
- Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Pediatric Hematology Oncology, Floating Hospital for Children at Tufts Medical Center, Boston, Massachusetts, United States of America
- Sackler School of Graduate Biomedical Sciences at Tufts University, Boston, Massachusetts, United States of America
- * E-mail: ,
| |
Collapse
|
36
|
Effect of ascorbate on plasminogen activator inhibitor-1 expression and release from platelets and endothelial cells in an in-vitro model of sepsis. Blood Coagul Fibrinolysis 2016; 26:436-42. [PMID: 25730478 DOI: 10.1097/mbc.0000000000000273] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The microcirculation during sepsis fails due to capillary plugging involving microthrombosis. We demonstrated that intravenous injection of ascorbate reduces this plugging, but the mechanism of this beneficial effect remains unclear. We hypothesize that ascorbate inhibits the release of the antifibrinolytic plasminogen activator inhibitor-1 (PAI-1) from endothelial cells and platelets during sepsis. Microvascular endothelial cells and platelets were isolated from mice. Cells were cultured and stimulated with lipopolysaccharide (LPS), tumor necrosis factor alpha (TNFα), or thrombin (agents of sepsis), with/without ascorbate for 1-24 h. PAI-1 mRNA was determined by quantitative PCR. PAI-1 protein release into the culture medium was measured by ELISA. In platelets, PAI-1 release was measured after LPS, TNFα, or thrombin stimulation, with/without ascorbate. In endothelial cells, LPS and TNFα increased PAI-1 mRNA after 6-24 h, but no increase in PAI-1 release was observed; ascorbate did not affect these responses. In platelets, thrombin, but not LPS or TNFα, increased PAI-1 release; ascorbate inhibited this increase at low extracellular pH. In unstimulated endothelial cells and platelets, PAI-1 is released into the extracellular space. Thrombin increases this release from platelets; ascorbate inhibits it pH-dependently. The data suggest that ascorbate promotes fibrinolysis in the microvasculature under acidotic conditions in sepsis.
Collapse
|
37
|
Mauler M, Seyfert J, Haenel D, Seeba H, Guenther J, Stallmann D, Schoenichen C, Hilgendorf I, Bode C, Ahrens I, Duerschmied D. Platelet-neutrophil complex formation-a detailed in vitro analysis of murine and human blood samples. J Leukoc Biol 2015; 99:781-9. [PMID: 26578648 DOI: 10.1189/jlb.3ta0315-082r] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/28/2015] [Indexed: 12/13/2022] Open
Abstract
Platelets form complexes with neutrophils during inflammatory processes. These aggregates migrate into affected tissues and also circulate within the organism. Several studies have evaluated platelet-neutrophil complexes as a marker of cardiovascular diseases in human and mouse. Although multiple publications have reported platelet-neutrophil complex counts, we noticed that different methods were used to analyze platelet-neutrophil complex formation, resulting in significant differences, even in baseline values. We established a protocol for platelet-neutrophil complex measurement with flow cytometry in murine and human whole blood samples. In vitro platelet-neutrophil complex formation was stimulated with ADP or PMA. We tested the effect of different sample preparation steps and cytometer settings on platelet-neutrophil complex detection and noticed false-positive counts with increasing acquisition speed. Platelet-neutrophil complex formation depends on platelet P-selectin expression, and antibody blocking of P-selectin consequently prevented ADP-induced platelet-neutrophil complex formation. These findings may help generating more comparable data among different research groups that examine platelet-neutrophil complexes as a marker for cardiovascular disease and novel therapeutic interventions.
Collapse
Affiliation(s)
- Maximilian Mauler
- Faculty of Biology, Heart Center, University of Freiburg, Germany and Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Julia Seyfert
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - David Haenel
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Hannah Seeba
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Janine Guenther
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Daniela Stallmann
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Claudia Schoenichen
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Ingo Ahrens
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Daniel Duerschmied
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| |
Collapse
|
38
|
Burnouf T, Chou ML, Goubran H, Cognasse F, Garraud O, Seghatchian J. An overview of the role of microparticles/microvesicles in blood components: Are they clinically beneficial or harmful? Transfus Apher Sci 2015; 53:137-45. [PMID: 26596959 DOI: 10.1016/j.transci.2015.10.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Blood cells and tissues generate heterogeneous populations of cell-derived vesicles, ranging from approximately 50 nm to 1 µm in diameter. Under normal physiological conditions and as an essential part of an energy-dependent natural process, microparticles (MPs) are continuously shed into the circulation from membranes of all viable cells such as megakaryocytes, platelets, red blood cells, white blood cells and endothelial cells. MP shedding can also be triggered by pathological activation of inflammatory processes and activation of coagulation or complement systems, or even by shear stress in the circulation. Structurally, MPs have a bilayered phospholipid structure exposing coagulant-active phosphatidylserine and expressing various membrane receptors, and they serve as cell-to-cell shuttles for bioactive molecules such as lipids, growth factors, microRNAs, and mitochondria. It was established that ex vivo processing of blood into its components, involving centrifugation, processing by various apheresis procedures, leucoreduction, pathogen reduction, and finally storage in different media and different types of blood bags, can impact MP generation and content. This is mostly due to exposure of the collected blood to anticoagulant/storage media and due to shear stresses or activation, contact with artificial surfaces, or exposure to various leucocyte-removal filters and pathogen-reduction treatments. Such artificially generated MPs, which are added to the original pool of MPs collected from the donor, may exhibit specific functional characteristics, as MPs are not an inert element of blood components. Not surprisingly, MPs' roles and functionality are therefore increasingly seen to be fully relevant to the field of transfusion medicine, and as a parameter of blood safety that must be considered in haemovigilance programmes. Continual advancements in assessment methods of MPs and storage lesions are gradually leading to a better understanding of the impacts of blood collection on MP generation, while clinical research should clarify links of MPs with transfusion reactions and certain clinical disorders. Harmonization and consensus in sampling protocols, sample handling and processing, and assessment methods are needed to achieve consensual interpretations. This review focuses on the role of MPs as an essential laboratory tool and as a most effective player in transfusion science and medicine and in health and disease.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| | - Ming-Li Chou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Loire, Saint-Etienne, France; GIMAP-EA3064, Université de Lyon, Saint Etienne, France
| | - Olivier Garraud
- Etablissement Français du Sang Auvergne-Loire, Saint-Etienne, France; Institut National de Transfusion Sanguine (INTS), Paris, France
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety, Audit/Inspection and DDR Strategy, London, UK.
| |
Collapse
|
39
|
Moore HB, Moore EE, Lawson PJ, Gonzalez E, Fragoso M, Morton AP, Gamboni F, Chapman MP, Sauaia A, Banerjee A, Silliman CC. Fibrinolysis shutdown phenotype masks changes in rodent coagulation in tissue injury versus hemorrhagic shock. Surgery 2015; 158:386-92. [PMID: 25979440 DOI: 10.1016/j.surg.2015.04.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/30/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Systemic hyperfibrinolysis (accelerated clot degradation) and fibrinolysis shutdown (impaired clot degradation) are associated with increased mortality compared with physiologic fibrinolysis after trauma. Animal models have not reproduced these changes. We hypothesize rodents have a shutdown phenotype that require an exogenous profibrinolytic to differentiate mechanisms that promote or inhibit fibrinolysis. METHODS Fibrinolysis resistance was assessed by thrombelastography (TEG) using exogenous tissue plasminogen activator (tPA) titrations in whole blood. There were 3 experimental groups: (1) tissue injury (laparotomy/bowel crush), (2) shock (hemorrhage to mean arterial pressure of 20 mmHg), and (3) control (arterial cannulation and tracheostomy). Baseline and 30-minute postintervention blood samples were collected, and assayed with TEG challenged with taurocholic acid (TUCA). RESULTS Rats were resistant to exogenous tPA; the percent clot remaining 30 minutes after maximum amplitude (CL30) at 150 ng/mL (P = .511) and 300 ng/mL (P = .931) was similar to baseline, whereas 600 ng/mL (P = .046) provoked fibrinolysis. Using the TUCA challenge, the percent change in CL30 from baseline was increased in tissue injury compared with control (P = .048.), whereas CL30 decreased in shock versus control (P = .048). tPA increased in the shock group compared with tissue injury (P = .009) and control (P = .012). CONCLUSION Rats have an innate fibrinolysis shutdown phenotype. The TEG TUCA challenge is capable of differentiating changes in clot stability with rats undergoing different procedures. Tissue injury inhibits fibrinolysis, whereas shock promotes tPA-mediated fibrinolysis.
Collapse
Affiliation(s)
- Hunter B Moore
- Denver Health Medical Center, Department of Surgery, Denver, CO; University of Colorado Denver, Department of Surgery, Denver, CO.
| | - Ernest E Moore
- Denver Health Medical Center, Department of Surgery, Denver, CO; University of Colorado Denver, Department of Surgery, Denver, CO
| | - Peter J Lawson
- Denver Health Medical Center, Department of Surgery, Denver, CO
| | - Eduardo Gonzalez
- Denver Health Medical Center, Department of Surgery, Denver, CO; University of Colorado Denver, Department of Surgery, Denver, CO
| | - Miguel Fragoso
- Denver Health Medical Center, Department of Surgery, Denver, CO
| | - Alex P Morton
- Denver Health Medical Center, Department of Surgery, Denver, CO; University of Colorado Denver, Department of Surgery, Denver, CO
| | - Fabia Gamboni
- University of Colorado Denver, Department of Surgery, Denver, CO
| | | | - Angela Sauaia
- University of Colorado Denver, Department of Surgery, Denver, CO
| | - Anirban Banerjee
- University of Colorado Denver, Department of Surgery, Denver, CO
| | - Christopher C Silliman
- University of Colorado Denver, Department of Surgery, Denver, CO; University of Colorado Denver, Department of Pediatrics, Denver, CO; Bonfils Blood Center, Denver, CO
| |
Collapse
|
40
|
Hårdstedt M, Lindblom S, Hong J, Nilsson B, Korsgren O, Ronquist G. A novel model for studies of blood-mediated long-term responses to cellular transplants. Ups J Med Sci 2015; 120:28-39. [PMID: 25322825 PMCID: PMC4389005 DOI: 10.3109/03009734.2014.965290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
AIMS Interaction between blood and bio-surfaces is important in many medical fields. With the aim of studying blood-mediated reactions to cellular transplants, we developed a whole-blood model for incubation of small volumes for up to 48 h. METHODS Heparinized polyvinyl chloride tubing was cut in suitable lengths and sealed to create small bags. Multiple bags, with fresh venous blood, were incubated attached to a rotating wheel at 37°C. Physiological variables in blood were monitored: glucose, blood gases, mono- and divalent cations and chloride ions, osmolality, coagulation (platelet consumption, thrombin-antithrombin complexes (TAT)), and complement activation (C3a and SC5b-9), haemolysis, and leukocyte viability. RESULTS Basic glucose consumption was high. Glucose depletion resulted in successive elevation of extracellular potassium, while sodium and calcium ions decreased due to inhibition of energy-requiring ion pumps. Addition of glucose improved ion balance but led to metabolic acidosis. To maintain a balanced physiological environment beyond 6 h, glucose and sodium hydrogen carbonate were added regularly based on analyses of glucose, pH, ions, and osmotic pressure. With these additives haemolysis was prevented for up to 72 h and leukocyte viability better preserved. Despite using non-heparinized blood, coagulation and complement activation were lower during long-term incubations compared with addition of thromboplastin and collagen. CONCLUSION A novel whole-blood model for studies of blood-mediated responses to a cellular transplant is presented allowing extended observations for up to 48 h and highlights the importance of stringent evaluations and adjustment of physiological conditions.
Collapse
Affiliation(s)
- Maria Hårdstedt
- Department of Immunology, Genetics and Pathology, Clinical Immunology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Center for Clinical Research Dalarna-Uppsala University, Falun, Sweden
| | - Susanne Lindblom
- Department of Immunology, Genetics and Pathology, Clinical Immunology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Jaan Hong
- Department of Immunology, Genetics and Pathology, Clinical Immunology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Clinical Immunology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Clinical Immunology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Gunnar Ronquist
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
41
|
Negrotto S, Jaquenod de Giusti C, Rivadeneyra L, Ure AE, Mena HA, Schattner M, Gomez RM. Platelets interact with Coxsackieviruses B and have a critical role in the pathogenesis of virus-induced myocarditis. J Thromb Haemost 2015; 13:271-82. [PMID: 25393316 DOI: 10.1111/jth.12782] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND To further understand the role of platelets in the pathogenesis of viral infections we explored platelet interaction with Coxsackieviruses B (CVB) 1 and 3. CVB is a group of viruses that cause the majority of human enterovirus-related viral myocarditis; their receptor (CAR) is expressed on the platelet surface and there is a well-characterized CVB3-induced myocarditis murine model. METHODS Human platelets were infected with CVB1 and 3 and viruses were detected in pellets and in supernatants. C57BL/6J mice with or without platelet depletion were inoculated with CVB3 and peripheral blood and heart samples collected at different times post-infection. RESULTS CVB1 and 3 RNA and a capsid protein were detected in infected platelets. Despite the fact that titration assays in Vero cells showed increasing infectivity titers over time, supernatants and pellets from infected platelets showed similar levels, suggesting that platelets were not susceptible to a replicative infectivity cycle. CVB binding was CAR-independent and resulted in P-selectin and phosphatidylserine (PS) exposure. CVB3-infected mice showed a rapid thrombocytopenia that correlated with an increase in platelet PS exposure and platelet-leukocyte aggregates without modification of platelet P-selectin expression or von Willebrand factor levels. Mortality, viremia, heart viral titers and myocarditis were significantly higher in platelet-depleted than normal animals. Type I IFN levels were not changed but IgG levels were lower in infected and platelet-depleted mice. CONCLUSIONS Our data reveal that platelets play a critical role in host survival and immune response against CVB3 infection.
Collapse
Affiliation(s)
- S Negrotto
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, ANM-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
42
|
Bottsford-Miller J, Choi HJ, Dalton HJ, Stone RL, Cho MS, Haemmerle M, Nick AM, Pradeep S, Zand B, Previs RA, Pecot CV, Crane EK, Hu W, Lutgendorf SK, Afshar-Kharghan V, Sood AK. Differential platelet levels affect response to taxane-based therapy in ovarian cancer. Clin Cancer Res 2014; 21:602-10. [PMID: 25473001 DOI: 10.1158/1078-0432.ccr-14-0870] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE We hypothesized that platelet levels during therapy could serve as a biomarker for response to therapy and that manipulation of platelet levels could impact responsiveness to chemotherapy. EXPERIMENTAL DESIGN The medical records of patients with recurrent or progressive ovarian cancer were retrospectively queried for changes in platelet and CA-125 levels during primary therapy. In vitro coculture experiments and in vivo orthotopic models of human ovarian cancer in mice were used to test the effect of modulating platelet levels on tumor growth and responsiveness to docetaxel. RESULTS Thrombocytosis at the diagnosis of ovarian cancer was correlated with decreased interval to progression (P = 0.05) and median overall survival (P = 0.007). Mean platelet levels corrected during primary therapy and rose at recurrence. Contrary to treatment-responsive patients, in a cohort of patients refractory to primary therapy, platelet levels did not normalize during therapy. In A2780, HeyA8, and SKOV3-ip1 ovarian cancer cell lines, platelet coculture protected against apoptosis (P < 0.05). In orthotopic models of human ovarian cancer, platelet depletion resulted in 70% reduced mean tumor weight (P < 0.05). Compared with mice treated with docetaxel, mice treated with both docetaxel and platelet-depleting antibody had a 62% decrease in mean tumor weight (P = 0.04). Platelet transfusion increased mean aggregate tumor weight 2.4-fold (P < 0.05), blocked the effect of docetaxel on tumor growth (P = 0.55) and decreased tumor cell apoptosis. Pretransfusion aspirinization of the platelets blocked the growth-promoting effects of transfusion. CONCLUSIONS Platelet-driven effects of chemotherapy response may explain clinical observations.
Collapse
Affiliation(s)
- Justin Bottsford-Miller
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hyun-Jin Choi
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather J Dalton
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca L Stone
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Min Soon Cho
- Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monika Haemmerle
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alpa M Nick
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunila Pradeep
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Behrouz Zand
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca A Previs
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chad V Pecot
- Department of Molecular Therapeutics, University of North Carolina Lineberger Comprehensive Cancer Center
| | - Erin King Crane
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Hu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan K Lutgendorf
- Department of Psychology, Obstetrics and Gynecology, and Urology, University of Iowa, Iowa City, Iowa
| | - Vahid Afshar-Kharghan
- Section of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
43
|
Riemann A, Ihling A, Thomas J, Schneider B, Thews O, Gekle M. Acidic environment activates inflammatory programs in fibroblasts via a cAMP-MAPK pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:299-307. [PMID: 25461841 DOI: 10.1016/j.bbamcr.2014.11.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/05/2014] [Accepted: 11/17/2014] [Indexed: 12/22/2022]
Abstract
The tissue micromilieu in disorders (inflammation, ischemia, tumor) often shows pronounced metabolic acidosis that may alter signaling and transcriptional activity in resident cells which can be of special importance for omnipresent fibroblasts. In the present study we investigated the impact of metabolic acidosis on rat fibroblasts with special emphasis on their role in inflammation by regulation of TNF-α, MCP-1, COX-2 and iNOS expression and the signaling pathways involved. Extracellular acidosis led to an enhanced expression of TNF-α, COX-2 and iNOS in parallel to an activation of p38 and ERK1/2 kinases that was not observed by sole intracellular acidosis. Accordingly, the protein amounts of TNF-α and COX-2 as well as the production of nitrate and nitrite were elevated. Acidosis-induced expression of COX-2 and iNOS depended on p38 kinase, but not on ERK1/2. In contrast acidosis-induced TNF-α expression was independent of both kinases. Although GPR4, GPR68 and GPR132 are expressed in fibroblasts, the involvement of these potential candidate pH sensors could be ruled out since no acidosis-induced elevation in intracellular cAMP or free calcium content was observed. Furthermore our data show that MAPK activation by an acidic micromilieu depends on Ser/Thr phosphatase activity, but not on the production of reactive oxygen species and is sensitive to cAMP antagonism by Rp-cAMPS. In conclusion, our results show that an acidic microenvironment induces a differential transcriptional program of pathological relevant genes in fibroblasts via the cAMP-phosphatase-MAPK pathway and thereby generates a parainflammatory situation that can result in tissue remodeling.
Collapse
Affiliation(s)
- A Riemann
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany.
| | - A Ihling
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany
| | - J Thomas
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany
| | - B Schneider
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany
| | - O Thews
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany
| | - M Gekle
- Julius-Bernstein-Institut für Physiologie, Universität Halle-Wittenberg, Germany
| |
Collapse
|
44
|
Clinically relevant concepts of haemostasis and detection of coagulopathy after trauma and surgery. ACTA ACUST UNITED AC 2014; 60:485-8. [PMID: 25374983 DOI: 10.1016/j.redar.2013.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Low level of procoagulant platelet microparticles is associated with impaired coagulation and transfusion requirements in trauma patients. J Trauma Acute Care Surg 2014; 77:692-700. [PMID: 25494419 DOI: 10.1097/ta.0000000000000437] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Following activation, platelets release small vesicles called platelet-derived microparticles (PMPs). PMPs accelerate thrombin generation and thus clot formation at sites of injury by exposing the procoagulant membrane phospholipid phosphatidylserine (PS). The role of PMPs in coagulopathy and hemorrhage following trauma remains elusive. We hypothesized that low levels of PS-positive PMPs (PS + PMPs) would be associated with impaired clot formation. METHODS This is a prospective observational study of 210 trauma patients admitted directly to a Level 1 trauma center. Plasma levels of PS + PMPs were determined by flow cytometry. Coagulation status was assessed by rotational thrombelastometry, and impaired clot formation was defined by an α angle less than 63 degrees using the tissue factor-based EXTEM reagent. Transfusion requirement was assessed by number of units of red blood cells (RBCs) transfused within 24 hours of admission; platelet aggregation capacity was evaluated by the Multiplate assay; and injury severity was determined by the Injury Severity Score (ISS). RESULTS The median ISS was 17, and blood samples were obtained after a median of 65 minutes following injury. Significantly lower levels of PS + PMPs were found in patients with impaired clot formation (p < 0.001). A low level of PS + PMPs was associated with a higher number of RBCs transfused during the initial 24 hours after admission (p < 0.03) when corrected for risk factors, for example, platelet count, hemoglobin level, and ISS. Platelet aggregation and PS + PMPs did not correlate significantly. CONCLUSION Low levels of PMPs were associated with impaired clot formation in trauma patients at admission and also with the number of RBC transfusions. This suggests that PMPs may play an important and not previously investigated role in trauma-induced coagulopathy. LEVEL OF EVIDENCE Prognostic study, level III.
Collapse
|
46
|
Etulain J, Fondevila C, Negrotto S, Schattner M. Platelet-mediated angiogenesis is independent of VEGF and fully inhibited by aspirin. Br J Pharmacol 2014; 170:255-65. [PMID: 23713888 DOI: 10.1111/bph.12250] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/08/2013] [Accepted: 05/17/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Platelets are major players in every step of vessel development through the local delivery of angiogenesis-modulating factors, including the pro-angiogenic protein VEGF and the anti-angiogenic endostatin. Although thrombin is a potent agonist and is highly elevated in angiogenesis-related diseases, studies regarding its action on the release of platelet angiogenic factors are scarce and controversial. Herein, we have investigated the role of thrombin not only in VEGF and endostatin release but also in net platelet angiogenic activity. EXPERIMENTAL APPROACH Human platelets were stimulated with thrombin in the presence of the various inhibitors of the signalling pathways involved in platelet activation. Supernatants/releasates were used to determine the levels of angiogenic molecules and to induce angiogenic responses. KEY RESULTS We found that thrombin induced the secretion of both VEGF and endostatin; however, the overall effect of the releasates was pro-angiogenic as they promoted tubule-like formation and increased the proliferation of endothelial cells. Both responses were only slightly suppressed in the presence of a VEGF receptor-neutralizing antibody. Pharmacological studies revealed that while inhibitors of PKC, p38, ERK1/2, Src kinases or PI3K/Akt exerted only partial inhibitory effects, aspirin fully blocked the pro-angiogenic activity of the releasate. CONCLUSIONS AND IMPLICATIONS In contrast to current belief, platelet pro-angiogenic responses are independent of VEGF and appear to be the result of the combined action of several molecules. Moreover, our data reinforce the notion that aspirin is a good candidate for a therapeutic agent to treat angiogenesis-related diseases.
Collapse
Affiliation(s)
- J Etulain
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
47
|
de Stoppelaar SF, van 't Veer C, van der Poll T. The role of platelets in sepsis. Thromb Haemost 2014; 112:666-77. [PMID: 24966015 DOI: 10.1160/th14-02-0126] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/16/2014] [Indexed: 01/01/2023]
Abstract
Platelets are small circulating anucleate cells that are of crucial importance in haemostasis. Over the last decade, it has become increasingly clear that platelets play an important role in inflammation and can influence both innate and adaptive immunity. Sepsis is a potentially lethal condition caused by detrimental host response to an invading pathogen. Dysbalanced immune response and activation of the coagulation system during sepsis are fundamental events leading to sepsis complications and organ failure. Platelets, being major effector cells in both haemostasis and inflammation, are involved in sepsis pathogenesis and contribute to sepsis complications. Platelets catalyse the development of hyperinflammation, disseminated intravascular coagulation and microthrombosis, and subsequently contribute to multiple organ failure. Inappropriate accumulation and activity of platelets are key events in the development of sepsis-related complications such as acute lung injury and acute kidney injury. Platelet activation readouts could serve as biomarkers for early sepsis recognition; inhibition of platelets in septic patients seems like an important target for immune-modulating therapy and appears promising based on animal models and retrospective human studies.
Collapse
Affiliation(s)
- Sacha F de Stoppelaar
- Sacha F. de Stoppelaar, MD, Academic Medical Centre, Centre of Experimental and Molecular Medicine, Meibergdreef 9, Room G2-130, 1105 AZ Amsterdam, the Netherlands, Tel.: +31 20 5665910, Fax: +31 20 6977192, E-mail:
| | | | | |
Collapse
|
48
|
Etulain J, Schattner M. Glycobiology of platelet-endothelial cell interactions. Glycobiology 2014; 24:1252-9. [DOI: 10.1093/glycob/cwu056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
49
|
Weng C, Li Y, Ryan P, Zhang Y, Liu F, Gao J, Bigger JT, Hripcsak G. A distribution-based method for assessing the differences between clinical trial target populations and patient populations in electronic health records. Appl Clin Inform 2014; 5:463-79. [PMID: 25024761 DOI: 10.4338/aci-2013-12-ra-0105] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/09/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To improve the transparency of clinical trial generalizability and to illustrate the method using Type 2 diabetes as an example. METHODS Our data included 1,761 diabetes clinical trials and the electronic health records (EHR) of 26,120 patients with Type 2 diabetes who visited Columbia University Medical Center of New-York Presbyterian Hospital. The two populations were compared using the Generalizability Index for Study Traits (GIST) on the earliest diagnosis age and the mean hemoglobin A1c (HbA1c) values. RESULTS Greater than 70% of Type 2 diabetes studies allow patients with HbA1c measures between 7 and 10.5, but less than 40% of studies allow HbA1c<7 and fewer than 45% of studies allow HbA1c>10.5. In the real-world population, only 38% of patients had HbA1c between 7 and 10.5, with 12% having values above the range and 52% having HbA1c<7. The GIST for HbA1c was 0.51. Most studies adopted broad age value ranges, with the most common restrictions excluding patients >80 or <18 years. Most of the real-world population fell within this range, but 2% of patients were <18 at time of first diagnosis and 8% were >80. The GIST for age was 0.75. CONCLUSIONS We contribute a scalable method to profile and compare aggregated clinical trial target populations with EHR patient populations. We demonstrate that Type 2 diabetes studies are more generalizable with regard to age than they are with regard to HbA1c. We found that the generalizability of age increased from Phase 1 to Phase 3 while the generalizability of HbA1c decreased during those same phases. This method can generalize to other medical conditions and other continuous or binary variables. We envision the potential use of EHR data for examining the generalizability of clinical trials and for defining population-representative clinical trial eligibility criteria.
Collapse
Affiliation(s)
- C Weng
- Department of Biomedical Informatics, Columbia University , New York, NY 10032
| | - Y Li
- Department of Computer Science, City College of New York , New York, NY 10031
| | - P Ryan
- Janssen Research and Development , Titusville, New Jersey, 08560 ; Observational Health Data Sciences and Informatics , New York, NY, 10032
| | - Y Zhang
- Department of Biostatistics, Columbia University , New York, NY 10032
| | - F Liu
- Department of Biomedical Informatics, Columbia University , New York, NY 10032
| | - J Gao
- Business School, Columbia University , New York, NY 10025
| | - J T Bigger
- Department of Medicine, Columbia University , New York, NY 10032
| | - G Hripcsak
- Department of Biomedical Informatics, Columbia University , New York, NY 10032
| |
Collapse
|
50
|
Önnheim K, Christenson K, Gabl M, Burbiel JC, Müller CE, Oprea TI, Bylund J, Dahlgren C, Forsman H. A novel receptor cross-talk between the ATP receptor P2Y2 and formyl peptide receptors reactivates desensitized neutrophils to produce superoxide. Exp Cell Res 2014; 323:209-217. [PMID: 24491917 DOI: 10.1016/j.yexcr.2014.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/19/2014] [Accepted: 01/22/2014] [Indexed: 11/24/2022]
Abstract
Neutrophils express several G-protein coupled receptors (GPCRs) and they cross regulate each other. We described a novel cross-talk mechanism in neutrophils, by which signals generated by the receptor for ATP (P2Y2) reactivate desensitized formyl peptide receptors (FPRs) so that these ligand-bound inactive FPRs resume signaling. At the signaling level, the cross-talk was unidirectional, i.e., P2Y2 ligation reactivated FPR, but not vice versa and was sensitive to the phosphatase inhibitor calyculinA. Further, we show that the cross talk between P2Y2 and FPR bypassed cytosolic Ca(2+) transients and did not rely on the actin cytoskeleton. In summary, our data demonstrate a novel cross-talk mechanism that results in reactivation of desensitized FPRs and, an amplification of the neutrophil response to ATP.
Collapse
Affiliation(s)
- Karin Önnheim
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden
| | - Karin Christenson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden
| | - Michael Gabl
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden
| | - Joachim C Burbiel
- PharmaCenter Bonn, Pharmaceutical Chemistry I at the Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Chemistry I at the Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Tudor I Oprea
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden; Division of Biocomputing, Department of Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Johan Bylund
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden
| | - Claes Dahlgren
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Göteborg 413 46, Sweden.
| |
Collapse
|