1
|
Blanco J, García A, Hermida‐Nogueira L, Castro AB. How to explain the beneficial effects of leukocyte- and platelet-rich fibrin. Periodontol 2000 2025; 97:74-94. [PMID: 38923566 PMCID: PMC11808445 DOI: 10.1111/prd.12570] [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: 01/24/2024] [Revised: 03/06/2024] [Accepted: 04/15/2024] [Indexed: 06/28/2024]
Abstract
The survival of an organism relies on its ability to repair the damage caused by trauma, toxic agents, and inflammation. This process involving cell proliferation and differentiation is driven by several growth factors and is critically dependent on the organization of the extracellular matrix. Since autologous platelet concentrates (APCs) are fibrin matrices in which cells, growth factors, and cytokines are trapped and delivered over time, they are able to influence that response at different levels. The present review thoroughly describes the molecular components present in one of these APCs, leukocyte- and platelet-rich fibrin (L-PRF), and summarizes the level of evidence regarding the influence of L-PRF on anti-inflammatory reactions, analgesia, hemostasis, antimicrobial capacity, and its biological mechanisms on bone/soft tissue regeneration.
Collapse
Affiliation(s)
- Juan Blanco
- Department of Surgery (Stomatology, Unit of Periodontology)Universidade de Santiago de CompostelaSantiago de CompostelaSpain
| | - Angel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS)Santiago de Compostela UniversitySantiago de CompostelaSpain
| | - Lidia Hermida‐Nogueira
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS)Santiago de Compostela UniversitySantiago de CompostelaSpain
| | - Ana B. Castro
- Department of Oral Health Sciences, Section of Periodontology, KU Leuven & DentistryUniversity Hospitals LeuvenLeuvenBelgium
| |
Collapse
|
2
|
Cai T, Burton M, McCafferty C, Van Den Helm S, Letunica N, Attard C, Horton S, Bottrell S, Schultz B, MacLaren G, Chiletti R, Best D, Johansen A, Newall F, Butt W, d'Udekem Y, Monagle P, Ignjatovic V. Immunofluorescence Protocol for Characterization of Platelet and Leukocyte Binding in Extracorporeal Membrane Oxygenation (ECMO) Circuits. ASAIO J 2025; 71:e1-e4. [PMID: 38905612 DOI: 10.1097/mat.0000000000002259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024] Open
Abstract
The continuous contact between blood and the foreign surface of the extracorporeal membrane oxygenation (ECMO) circuit contributes to hemostatic, inflammatory, and other physiological disturbances observed during ECMO. Although previous studies have extensively investigated blood samples from patients on ECMO, cell adsorption to the ECMO circuit as an additional factor that could potentially influence clinical outcomes, has largely been overlooked. Here we provide a detailed immunofluorescence (IF) protocol designed to characterize cellular binding on ECMO circuits collected from patients. Extracorporeal membrane oxygenation circuits were collected from three pediatric patients and an albumin primed-only ECMO circuit was used as control. Circuit samples from five different sites within each ECMO circuit were collected and processed for the IF protocol. CD14 and CD42a antibodies were used to identify platelets and leukocytes bound to each ECMO circuit sample and images captured using inverted fluorescence microscopy. The protocol enables the comprehensive characterization of platelet and leukocyte binding to ECMO circuits collected from patients, which could in turn extend our knowledge of the characteristics of circuit binding and may provide guidance for improved ECMO circuit design.
Collapse
Affiliation(s)
- Tengyi Cai
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Matthew Burton
- Flow Cytometry and Imaging, Murdoch Children's Research Institute, Parkville, Australia
| | - Conor McCafferty
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Suelyn Van Den Helm
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Natasha Letunica
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
| | - Chantal Attard
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Stephen Horton
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Cardiac Surgery, The Royal Children's Hospital, Parkville, Australia
| | - Steve Bottrell
- Cardiac Surgery, The Royal Children's Hospital, Parkville, Australia
| | - Bradley Schultz
- Cardiac Surgery, The Royal Children's Hospital, Parkville, Australia
| | - Graeme MacLaren
- Cardiothoracic Intensive Care Unit, National University Health System, Singapore
| | - Roberto Chiletti
- Intensive Care, The Royal Children's Hospital, Parkville, Australia
- Paediatric Intensive Care Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Derek Best
- Intensive Care, The Royal Children's Hospital, Parkville, Australia
- Paediatric Intensive Care Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Amy Johansen
- Intensive Care, The Royal Children's Hospital, Parkville, Australia
- Paediatric Intensive Care Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Fiona Newall
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Clinical Haematology, The Royal Children's Hospital, Parkville, Australia
| | - Warwick Butt
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Intensive Care, The Royal Children's Hospital, Parkville, Australia
- Paediatric Intensive Care Research Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Yves d'Udekem
- Cardiovascular Surgery, Children's National Heart Institute, Washington, DC
| | - Paul Monagle
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Clinical Haematology, The Royal Children's Hospital, Parkville, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Vera Ignjatovic
- From the Haematology Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Cardiac Surgery, The Royal Children's Hospital, Parkville, Australia
- Johns Hopkins All Children's Institute for Clinical and Translational Research, St Petersburg, Florida
- Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
3
|
Garcia T, Petrera A, Hauck SM, Baber R, Wirkner K, Kirsten H, Pott J, Tönjes A, Henger S, Loeffler M, Peters A, Scholz M. Relationship of proteins and subclinical cardiovascular traits in the population-based LIFE-Adult study. Atherosclerosis 2024; 398:118613. [PMID: 39340936 DOI: 10.1016/j.atherosclerosis.2024.118613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND AND AIMS Understanding molecular processes of the early phase of atherosclerotic cardiovascular disease conditions is of utmost importance for early prediction and intervention measures. METHODS We measured 92 cardiovascular-disease-related proteins (Olink, Cardiovascular III) in 2024 elderly participants of the population-based LIFE-Adult study. We analysed the impact of 27 covariables on these proteins including blood counts, cardiovascular risk factors and life-style-related parameters. We also analysed protein associations with 13 subclinical cardiovascular traits comprising carotid intima media thickness, plaque burden, three modes of Vicorder-based pulse-wave velocities, ankle-brachial index and ECLIA-based N-terminal prohormone of brain natriuretic peptide (NT-proBNP). RESULTS Estimated glomerular filtration rate, triglycerides and sex where the most relevant covariables explaining more than 1 % variance of 49, 22 and 20 proteins, respectively. A total of 43 proteins were significantly associated with at least one of the analysed subclinical cardiovascular traits. NT-pro-BNP, brachial-ankle pulse-wave velocity (baPWV) and parameters of carotid plaque burden accounted for the largest number of associations. Association overlaps were relatively sparse. Only growth/differentiation factor 15, low density lipoprotein receptor and interleukin-1 receptor type 2 are associated with these three different cardiovascular traits. We confirmed several literature findings and found yet unreported associations for carotid plaque presence (von-Willebrand factor, galectin 4), carotid intima-media thickness (carboxypeptidase A1 andB1), baPWV (cathepsin D) and NT-proBNP (cathepsin Z, low density lipoprotein receptor, neurogenic locus homolog protein 3, trem-like transcript 2). Sex-interaction effects were observed, e.g. for spondin-1 and growth/differentiation factor 15 likely regulated by androgen response elements. CONCLUSIONS We extend the catalogue of proteome biomarkers possibly involved in early stages of cardiovascular disease pathologies providing targets for early risk prediction or intervention strategies.
Collapse
Affiliation(s)
- Tarcyane Garcia
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Ronny Baber
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Wirkner
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Anke Tönjes
- Department of Medicine, Division of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Sylvia Henger
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany; Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
4
|
Strelkova OS, Osgood RT, Tian C, Zhang X, Hale E, De-la-Torre P, Hathaway DM, Indzhykulian AA. PKHD1L1 is required for stereocilia bundle maintenance, durable hearing function and resilience to noise exposure. Commun Biol 2024; 7:1423. [PMID: 39482437 PMCID: PMC11527881 DOI: 10.1038/s42003-024-07121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Polycystic Kidney and Hepatic Disease 1-Like 1 (PKHD1L1) is a human deafness gene, responsible for autosomal recessive deafness-124 (DFNB124). Sensory hair cells of the cochlea are essential for hearing, relying on the mechanosensitive stereocilia bundle at their apical pole for their function. PKHD1L1 is a stereocilia protein required for the formation of the developmentally transient stereocilia surface coat. In this study, we carry out an in depth characterization of PKHD1L1 expression in mice during development and adulthood, analyze hair-cell bundle morphology and hearing function in aging PKHD1L1-deficient mouse lines, and assess their susceptibility to noise damage. Our findings reveal that PKHD1L1-deficient mice display no disruption to bundle cohesion or tectorial membrane attachment-crown formation during development. However, starting from 6 weeks of age, PKHD1L1-deficient mice display missing stereocilia and disruptions to bundle coherence. Both conditional and constitutive PKHD1L1 knockout mice develop high-frequency hearing loss progressing to lower frequencies with age. Furthermore, PKHD1L1-deficient mice are susceptible to permanent hearing loss following moderate acoustic overexposure, which induces only temporary hearing threshold shifts in wild-type mice. These results suggest a role for PKHD1L1 in establishing robust sensory hair bundles during development, necessary for maintaining bundle cohesion and function in response to acoustic trauma and aging.
Collapse
Affiliation(s)
- Olga S Strelkova
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Richard T Osgood
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Chunjie Tian
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Xinyuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Evan Hale
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Pedro De-la-Torre
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Daniel M Hathaway
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Artur A Indzhykulian
- Department of Otolaryngology Head and Neck Surgery, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Debaene C, Feys HB, Six KR. Shedding light on GPIbα shedding. Curr Opin Hematol 2024; 31:224-229. [PMID: 38728102 DOI: 10.1097/moh.0000000000000826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW Ectodomain shedding has been investigated since the late 1980s. The abundant and platelet specific GPIbα receptor is cleaved by ADAM17 resulting in the release of its ectodomain called glycocalicin. This review will address the role of glycocalicin as an end-stage marker of platelet turnover and storage lesion and will consider a potential function as effector in processes beyond hemostasis. RECENT FINDINGS Glycocalicin has been described as a marker for platelet senescence, turnover and storage lesion but is not routinely used in a clinical setting because its diagnostic value is nondiscriminatory. Inhibition of glycocalicin shedding improves posttransfusion recovery but little is known (yet) about potential hemostatic improvements. In physiological settings, GPIbα shedding is restricted to the intracellular GPIbα receptor subpopulation suggesting a role for shedding or glycocalicin beyond hemostasis. SUMMARY So far, all evidence represents glycocalicin as an end-stage biomarker of platelet senescence and a potential trigger for platelet clearance. The extensive list of interaction partners of GPIbα in fields beyond hemostasis opens new possibilities to investigate specific effector functions of glycocalicin.
Collapse
Affiliation(s)
- Caitlin Debaene
- Transfusion Research Center, Belgian Red Cross Flanders
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross Flanders
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Katrijn R Six
- Transfusion Research Center, Belgian Red Cross Flanders
| |
Collapse
|
6
|
Granja T, Köhler D, Tang L, Burkard P, Eggstein C, Hemmen K, Heinze KG, Heck-Swain KL, Koeppen M, Günther S, Blaha M, Magunia H, Bamberg M, Konrad F, Ngamsri KC, Fuhr A, Keller M, Bernard AM, Haeberle HA, Bakchoul T, Zarbock A, Nieswandt B, Rosenberger P. Semaphorin 7A coordinates neutrophil response during pulmonary inflammation and sepsis. Blood Adv 2024; 8:2660-2674. [PMID: 38489236 PMCID: PMC11157222 DOI: 10.1182/bloodadvances.2023011778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024] Open
Abstract
ABSTRACT Pulmonary defense mechanisms are critical for host integrity during pneumonia and sepsis. This defense is fundamentally dependent on the activation of neutrophils during the innate immune response. Recent work has shown that semaphorin 7A (Sema7A) holds significant impact on platelet function, yet its role on neutrophil function within the lung is not well understood. This study aimed to identify the role of Sema7A during pulmonary inflammation and sepsis. In patients with acute respiratory distress syndrome (ARDS), we were able to show a correlation between Sema7A and oxygenation levels. During subsequent workup, we found that Sema7A binds to the neutrophil PlexinC1 receptor, increasing integrins, and L-selectin on neutrophils. Sema7A prompted neutrophil chemotaxis in vitro and the formation of platelet-neutrophil complexes in vivo. We also observed altered adhesion and transmigration of neutrophils in Sema7A-/-animals in the lung during pulmonary inflammation. This effect resulted in increased number of neutrophils in the interstitial space of Sema7A-/- animals but reduced numbers of neutrophils in the alveolar space during pulmonary sepsis. This finding was associated with significantly worse outcome of Sema7A-/- animals in a model of pulmonary sepsis. Sema7A has an immunomodulatory effect in the lung, affecting pulmonary sepsis and ARDS. This effect influences the response of neutrophils to external aggression and might influence patient outcome. This trial was registered at www.ClinicalTrials.gov as #NCT02692118.
Collapse
Affiliation(s)
- Tiago Granja
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
- Lusofona's Research Center for Biosciences & Health Technologies, CBIOS-Universidade, Lisboa, Portugal
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Linyan Tang
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
- Department of Intensive Care Medicine, Shenzhen University General Hospital, Shenzhen, China
| | - Philipp Burkard
- Institute of Experimental Biomedicine, University Hospital, University of Würzburg, Würzburg, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Katherina Hemmen
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Katrin G. Heinze
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Ka-Lin Heck-Swain
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Michael Koeppen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Sven Günther
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Maximilian Blaha
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Harry Magunia
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Maximilian Bamberg
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Franziska Konrad
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | | | - Anika Fuhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Marius Keller
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Alice M. Bernard
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Helene A. Haeberle
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Tamam Bakchoul
- Center for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Alexander Zarbock
- Department of Anesthesiology and Intensive Care Medicine and Pain Medicine, University Hospital, Münster, Germany
| | - Bernhard Nieswandt
- Department of Intensive Care Medicine, Shenzhen University General Hospital, Shenzhen, China
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| |
Collapse
|
7
|
Redfield SE, De-la-Torre P, Zamani M, Wang H, Khan H, Morris T, Shariati G, Karimi M, Kenna MA, Seo GH, Xu H, Lu W, Naz S, Galehdari H, Indzhykulian AA, Shearer AE, Vona B. PKHD1L1, a gene involved in the stereocilia coat, causes autosomal recessive nonsyndromic hearing loss. Hum Genet 2024; 143:311-329. [PMID: 38459354 PMCID: PMC11043200 DOI: 10.1007/s00439-024-02649-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/21/2024] [Indexed: 03/10/2024]
Abstract
Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modeling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modeling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.
Collapse
Affiliation(s)
- Shelby E Redfield
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA, 02115, USA
| | - Pedro De-la-Torre
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, MA, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| | - Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
| | - Hanjun Wang
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, No. 40 Daxuebei Road, Zhengzhou, 450052, China
| | - Hina Khan
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Tyler Morris
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, MA, USA
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Karimi
- Khuzestan Cochlear Implantation Center (Tabassom), Ahvaz, Iran
| | - Margaret A Kenna
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA, 02115, USA
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, MA, USA
| | | | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, No. 40 Daxuebei Road, Zhengzhou, 450052, China
| | - Wei Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian-She Road, Zhengzhou, 450052, China
| | - Sadaf Naz
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Hamid Galehdari
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Artur A Indzhykulian
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, MA, USA.
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA.
| | - A Eliot Shearer
- Department of Otolaryngology and Communication Enhancement, Boston Children's Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA, 02115, USA.
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA.
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, 37073, Göttingen, Germany.
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075, Göttingen, Germany.
| |
Collapse
|
8
|
Redfield SE, De-la-Torre P, Zamani M, Wang H, Khan H, Morris T, Shariati G, Karimi M, Kenna MA, Seo GH, Xu H, Lu W, Naz S, Galehdari H, Indzhykulian AA, Shearer AE, Vona B. PKHD1L1, A Gene Involved in the Stereocilia Coat, Causes Autosomal Recessive Nonsyndromic Hearing Loss. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.08.23296081. [PMID: 37873491 PMCID: PMC10593026 DOI: 10.1101/2023.10.08.23296081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modelling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modelling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.
Collapse
Affiliation(s)
- Shelby E. Redfield
- Department of Otolaryngology and Communication Enhancement, Boston Children’s Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA 02115, USA
| | - Pedro De-la-Torre
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, Massachusetts, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
| | - Hanjun Wang
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, No. 40 Daxuebei Road, Zhengzhou, 450052, China
| | - Hina Khan
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore 54590, Pakistan
| | - Tyler Morris
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, Massachusetts, USA
| | - Gholamreza Shariati
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz, Iran
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Karimi
- Khuzestan Cochlear Implantation Center (Tabassom), Ahvaz, Iran
| | - Margaret A. Kenna
- Department of Otolaryngology and Communication Enhancement, Boston Children’s Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA 02115, USA
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, Massachusetts, USA
| | | | - Hongen Xu
- Precision Medicine Center, Academy of Medical Science, Zhengzhou University, No. 40 Daxuebei Road, Zhengzhou, 450052, China
| | - Wei Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jian-she Road, Zhengzhou, 450052, China
| | - Sadaf Naz
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore 54590, Pakistan
| | - Hamid Galehdari
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Artur A. Indzhykulian
- Mass Eye and Ear, Eaton Peabody Laboratories, Boston, Massachusetts, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - A. Eliot Shearer
- Department of Otolaryngology and Communication Enhancement, Boston Children’s Hospital, 300 Longwood Avenue, BCH-3129, Boston, MA 02115, USA
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
9
|
Lee CSM, Kaur A, Montague SJ, Hicks SM, Andrews RK, Gardiner EE. Tissue inhibitors of metalloproteinases (TIMPs) modulate platelet ADAM10 activity. Platelets 2023; 34:2288213. [PMID: 38031964 DOI: 10.1080/09537104.2023.2288213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Platelet-specific collagen receptor glycoprotein (GP)VI is stable on the surface of circulating platelets but undergoes ectodomain cleavage on activated platelets. Activation-dependent GPVI metalloproteolysis is primarily mediated by A Disintegrin And Metalloproteinase (ADAM) 10. Regulation of platelet ADAMs activity is not well-defined however Tissue Inhibitors of Metalloproteinases (TIMPs) may play a role. As levels of TIMPs on platelets and the control of ADAMs-mediated shedding by TIMPs has not been evaluated, we quantified the levels of TIMPs on the surface of resting and activated platelets from healthy donors by flow cytometry and multiplex ELISA. Variable levels of all TIMPs could be detected on platelets. Plasma contained significant quantities of TIMP1 and TIMP2, but only trace amounts of TIMP3 and TIMP4. Recombinant TIMP3 strongly ablated resting and activated platelet ADAM10 activity, when monitored using a quenched fluorogenic peptide substrate with ADAM10 specificity. Whilst ADAM10-specific inhibitor GI254023X or ethylenediamine tetraacetic acid (EDTA) could modulate ligand-initiated shedding of GPVI, only recombinant TIMP2 achieved a modest (~20%) inhibition. We conclude that some platelet TIMPs are able to modulate platelet ADAM10 activity but none strongly regulate ligand-dependent shedding of GPVI. Our findings provide new insights into the regulation of platelet receptor sheddase activity.
Collapse
Affiliation(s)
- Christine Shu Mei Lee
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Amandeep Kaur
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Samantha J Montague
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah M Hicks
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Robert K Andrews
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
10
|
Bayrón-Marrero Z, Branfield S, Menéndez-Pérez J, Nieves-López B, Ospina L, Cantres-Rosario Y, Melendez LM, Hunter R, Gibson A, Maldonado-Martínez G, Washington AV. The Characterization and Evaluation of the Soluble Triggering Receptor Expressed on Myeloid Cells-like Transcript-1 in Stable Coronary Artery Disease. Int J Mol Sci 2023; 24:13632. [PMID: 37686440 PMCID: PMC10487797 DOI: 10.3390/ijms241713632] [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: 08/06/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Platelets play crucial roles in the development and progression of coronary artery disease (CAD). The triggering receptor expressed in myeloid cells-like transcript-1 (TLT-1) is stored in platelet α granules, and activated platelets release a soluble fragment (sTLT-1). We set out to better characterize the constituent amino acids of sTLT-1 and to evaluate sTLT-1 for use as a biomarker in patients with stable CAD. We evaluated sTLT-1 release using immunoprecipitation and mass spectrometry and employed statistical methods to retrospectively correlate sTLT-1 concentrations, utilizing ELISA in plasma samples from 1510 patients with documented stable CAD. We identified TLT-1 residues to 133 in platelet releasates. ADAM17 cuts TLT-1, suggesting that S136 is the C-terminal amino acid in sTLT-1. Our results revealed that for CAD patients, sTLT-1 levels did not differ significantly according to primary outcomes of death or major cardiac event; however, patients with left ventricular (LV) dysfunction had significantly lower plasma sTLT-1 levels as compared to those with normal LV function (981.62 ± 1141 pg/mL vs. 1247.48 ± 1589 pg/mL; p = 0.003). When patients were stratified based on sTLT-1 peak frequency distribution (544 pg/mL), a significant association with congestive heart failure was identified (OR = 2.94; 1.040-8.282; p = 0.042), which could be explained by LV dysfunction.
Collapse
Affiliation(s)
- Zaida Bayrón-Marrero
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
| | - Siobhan Branfield
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
- Department of Biology, Oakland University, Rochester Hills, MI 48309, USA
| | - Javier Menéndez-Pérez
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
| | - Benjamín Nieves-López
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
| | - Laura Ospina
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
| | - Yadira Cantres-Rosario
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (Y.C.-R.); (L.M.M.)
| | - Loyda M. Melendez
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, San Juan, PR 00936, USA; (Y.C.-R.); (L.M.M.)
- Department of Microbiology and Medical Zoology, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA
| | - Robert Hunter
- Retroviral Research Center, Universidad Central del Caribe, Bayamón, PR 00960, USA;
| | - Angelia Gibson
- Division of Natural Sciences, Maryville College, Maryville, TN 37804, USA;
| | | | - A. Valance Washington
- Department of Biology, University of Puerto Rico–Rio Piedras, San Juan, PR 00936, USA; (Z.B.-M.); (S.B.); (J.M.-P.); (B.N.-L.); (L.O.)
- Department of Biology, Oakland University, Rochester Hills, MI 48309, USA
- William Beaumont School of Medicine, Oakland University, Rochester Hills, MI 48309, USA
| |
Collapse
|
11
|
Amin A, Badenes M, Tüshaus J, de Carvalho É, Burbridge E, Faísca P, Trávníčková K, Barros A, Carobbio S, Domingos PM, Vidal-Puig A, Moita LF, Maguire S, Stříšovský K, Ortega FJ, Fernández-Real JM, Lichtenthaler SF, Adrain C. Semaphorin 4B is an ADAM17-cleaved adipokine that inhibits adipocyte differentiation and thermogenesis. Mol Metab 2023; 73:101731. [PMID: 37121509 PMCID: PMC10197113 DOI: 10.1016/j.molmet.2023.101731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023] Open
Abstract
OBJECTIVE The metalloprotease ADAM17 (also called TACE) plays fundamental roles in homeostasis by shedding key signaling molecules from the cell surface. Although its importance for the immune system and epithelial tissues is well-documented, little is known about the role of ADAM17 in metabolic homeostasis. The purpose of this study was to determine the impact of ADAM17 expression, specifically in adipose tissues, on metabolic homeostasis. METHODS We used histopathology, molecular, proteomic, transcriptomic, in vivo integrative physiological and ex vivo biochemical approaches to determine the impact of adipose tissue-specific deletion of ADAM17 upon adipocyte and whole organism metabolic physiology. RESULTS ADAM17adipoq-creΔ/Δ mice exhibited a hypermetabolic phenotype characterized by elevated energy consumption and increased levels of adipocyte thermogenic gene expression. On a high fat diet, these mice were more thermogenic, while exhibiting elevated expression levels of genes associated with lipid oxidation and lipolysis. This hypermetabolic phenotype protected mutant mice from obesogenic challenge, limiting weight gain, hepatosteatosis and insulin resistance. Activation of beta-adrenoceptors by the neurotransmitter norepinephrine, a key regulator of adipocyte physiology, triggered the shedding of ADAM17 substrates, and regulated ADAM17 expression at the mRNA and protein levels, hence identifying a functional connection between thermogenic licensing and the regulation of ADAM17. Proteomic studies identified Semaphorin 4B (SEMA4B), as a novel ADAM17-shed adipokine, whose expression is regulated by physiological thermogenic cues, that acts to inhibit adipocyte differentiation and dampen thermogenic responses in adipocytes. Transcriptomic data showed that cleaved SEMA4B acts in an autocrine manner in brown adipocytes to repress the expression of genes involved in adipogenesis, thermogenesis, and lipid uptake, storage and catabolism. CONCLUSIONS Our findings identify a novel ADAM17-dependent axis, regulated by beta-adrenoceptors and mediated by the ADAM17-cleaved form of SEMA4B, that modulates energy balance in adipocytes by inhibiting adipocyte differentiation, thermogenesis and lipid catabolism.
Collapse
Affiliation(s)
- Abdulbasit Amin
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | - Marina Badenes
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Faculty of Veterinary Medicine, Lusofona University, Lisbon, Portugal; Faculty of Veterinary Nursing, Polytechnic Institute of Lusofonia, Lisbon, Portugal
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Érika de Carvalho
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Emma Burbridge
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Pedro Faísca
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Květa Trávníčková
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - André Barros
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Stefania Carobbio
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Pedro M Domingos
- Instituto de Tecnologia Química da Universidade Nova de Lisboa (ITQB-Nova), Oeiras, Portugal
| | - Antonio Vidal-Puig
- Centro de Investigacíon Principe Felipe (CIPF), Valencia, Spain; Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, UK
| | - Luís F Moita
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Sarah Maguire
- Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland
| | - Kvido Stříšovský
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Francisco J Ortega
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - José Manuel Fernández-Real
- Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Department of Medical Sciences, University of Girona, Girona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), and Institute of Salud Carlos III (ISCIII), Madrid, Spain
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Colin Adrain
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal; Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, N. Ireland.
| |
Collapse
|
12
|
Trivigno SMG, Guidetti GF, Barbieri SS, Zarà M. Blood Platelets in Infection: The Multiple Roles of the Platelet Signalling Machinery. Int J Mol Sci 2023; 24:ijms24087462. [PMID: 37108623 PMCID: PMC10138547 DOI: 10.3390/ijms24087462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Platelets are classically recognized for their important role in hemostasis and thrombosis but they are also involved in many other physiological and pathophysiological processes, including infection. Platelets are among the first cells recruited to sites of inflammation and infection and they exert their antimicrobial response actively cooperating with the immune system. This review aims to summarize the current knowledge on platelet receptor interaction with different types of pathogens and the consequent modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Silvia M G Trivigno
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- University School for Advanced Studies, IUSS, 27100 Pavia, Italy
| | | | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| |
Collapse
|
13
|
Morris SM, Chauhan A. The role of platelet mediated thromboinflammation in acute liver injury. Front Immunol 2022; 13:1037645. [PMID: 36389830 PMCID: PMC9647048 DOI: 10.3389/fimmu.2022.1037645] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022] Open
Abstract
Acute liver injuries have wide and varied etiologies and they occur both in patients with and without pre-existent chronic liver disease. Whilst the pathophysiological mechanisms remain distinct, both acute and acute-on-chronic liver injury is typified by deranged serum transaminase levels and if severe or persistent can result in liver failure manifest by a combination of jaundice, coagulopathy and encephalopathy. It is well established that platelets exhibit diverse functions as immune cells and are active participants in inflammation through processes including immunothrombosis or thromboinflammation. Growing evidence suggests platelets play a dualistic role in liver inflammation, shaping the immune response through direct interactions and release of soluble mediators modulating function of liver sinusoidal endothelial cells, stromal cells as well as migrating and tissue-resident leucocytes. Elucidating the pathways involved in initiation, propagation and resolution of the immune response are of interest to identify therapeutic targets. In this review the provocative role of platelets is outlined, highlighting beneficial and detrimental effects in a spatial, temporal and disease-specific manner.
Collapse
Affiliation(s)
- Sean M. Morris
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Abhishek Chauhan
- The Liver Unit, University Hospitals Birmingham, Birmingham, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Abhishek Chauhan,
| |
Collapse
|
14
|
Kubo M, Sakai K, Hayakawa M, Kashiwagi H, Yagi H, Seki Y, Hasegawa A, Tanaka H, Amano I, Tomiyama Y, Matsumoto M. Increased cleavage of von Willebrand factor by ADAMTS13 may contribute strongly to acquired von Willebrand syndrome development in patients with essential thrombocythemia. J Thromb Haemost 2022; 20:1589-1598. [PMID: 35352474 DOI: 10.1111/jth.15717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Patients with essential thrombocythemia (ET) often experience bleeding associated with acquired von Willebrand syndrome (AVWS) when the platelet count is markedly increased. OBJECTIVE We investigated whether von Willebrand factor (VWF) degradation is enhanced in patients with ET. METHODS Seventy patients with ET underwent VWF multimer (VWFM) analysis and measurement of VWF-related parameters. We calculated the VWFM index, defined as the ratio of intensities of a patient's molecular weight-categorized VWFMs, and those of a healthy subject's, using densitometric analysis. VWF degradation product (DP) was measured via ELISA using a monoclonal antibody that specifically recognizes Y1605 at the C-terminal boundary, which is exposed following ADAMTS13-mediated cleavage of the Y1605-M1606 bond of the VWF A2 domain. RESULTS Patients with higher platelet counts had a significantly reduced high molecular weight (HMW)-VWFM index and an increased VWF-DP:VWF antigen (Ag) ratio compared to those with lower platelet counts. On multivariate analysis, the VWF-DP/VWF:Ag ratio was an independent predictor of the HMW-VWFM index. Patients who underwent cytoreductive therapy had a significantly higher HMW-VWFM index and lower VWF-DP/VWF:Ag ratio than those who did not. Among individual patients, there was also a significant increase in the HMW-VWFM index and a decrease in the VWF-DP/VWF:Ag ratio after cytoreductive therapy compared to pre-therapy values. CONCLUSION In patients with ET, an increased platelet count is associated with enhanced cleavage of VWF at the Y1605-M1606 bond, primarily by ADAMTS13, leading to AVWS. Cytoreductive therapy reduces the platelet count, prevents excessive VWF cleavage, and improves VWFM distributions.
Collapse
Affiliation(s)
- Masayuki Kubo
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara, Japan
- Department of Hematology, Nara Medical University, Kashihara, Japan
| | - Kazuya Sakai
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara, Japan
| | - Masaki Hayakawa
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara, Japan
| | - Hirokazu Kashiwagi
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Blood Transfusion, Osaka University Hospital, Suita, Japan
| | - Hideo Yagi
- Department of Hematology and Oncology, Nara Prefecture General Medical Center, Nara, Japan
| | - Yoshinobu Seki
- Department of Hematology, Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, Minamiuonuma, Japan
| | - Atsushi Hasegawa
- Department of Hematology, Nara Medical University, Kashihara, Japan
| | - Haruyuki Tanaka
- Department of Hematology, Nara Medical University, Kashihara, Japan
| | - Itsuto Amano
- Department of Hematology, Nara Medical University, Kashihara, Japan
| | - Yoshiaki Tomiyama
- Department of Blood Transfusion, Osaka University Hospital, Suita, Japan
| | - Masanori Matsumoto
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara, Japan
| |
Collapse
|
15
|
Napolitano F, Montuori N. Role of Plasminogen Activation System in Platelet Pathophysiology: Emerging Concepts for Translational Applications. Int J Mol Sci 2022; 23:ijms23116065. [PMID: 35682744 PMCID: PMC9181697 DOI: 10.3390/ijms23116065] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Traditionally, platelets have been exclusively considered for their procoagulant and antifibrinolytic effects during normal activation of hemostasis. Effectively, activated platelets secrete coagulation factors, expose phosphatidylserine, and promote thrombin and fibrin production. In addition to procoagulant activities, platelets confer resistance of thrombi to fibrinolysis by inducing clot retraction of the fibrin network and release of huge amounts of plasminogen activator inhibitor-1, which is the major physiologic inhibitor of the fibrinolytic cascade. However, the discovery of multiple relations with the fibrinolytic system, also termed Plasminogen Activation System (PAS), has introduced new perspectives on the platelet role in fibrinolysis. Indeed, the activated membrane surface of platelets provides binding sites on which fibrinolytic enzymes can be activated. This review discusses the evidence of the profibrinolytic properties of platelets through the description of PAS components and related proteins that are contained in or bind to platelets. Our analyses of literature data lead to the conclusion that in the initial phase of the hemostatic process, antifibrinolytic effects prevail over profibrinolytic activity, but at later stages, platelets might enhance fibrinolysis through the engagement of PAS components. A better understanding of spatial and temporal characteristics of platelet-mediated fibrinolysis during normal hemostasis could improve therapeutic options for bleeding and thrombotic disorders.
Collapse
|
16
|
Tyagi T, Jain K, Gu SX, Qiu M, Gu VW, Melchinger H, Rinder H, Martin KA, Gardiner EE, Lee AI, Ho Tang W, Hwa J. A guide to molecular and functional investigations of platelets to bridge basic and clinical sciences. NATURE CARDIOVASCULAR RESEARCH 2022; 1:223-237. [PMID: 37502132 PMCID: PMC10373053 DOI: 10.1038/s44161-022-00021-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/17/2022] [Indexed: 07/29/2023]
Abstract
Platelets have been shown to be associated with pathophysiological process beyond thrombosis, demonstrating critical additional roles in homeostatic processes, such as immune regulation, and vascular remodeling. Platelets themselves can have multiple functional states and can communicate and regulate other cells including immune cells and vascular smooth muscle cells, to serve such diverse functions. Although traditional platelet functional assays are informative and reliable, they are limited in their ability to unravel platelet phenotypic heterogeneity and interactions. Developments in methods such as electron microscopy, flow cytometry, mass spectrometry, and 'omics' studies, have led to new insights. In this Review, we focus on advances in platelet biology and function, with an emphasis on current and promising methodologies. We also discuss technical and biological challenges in platelet investigations. Using coronavirus disease 2019 (COVID-19) as an example, we further describe the translational relevance of these approaches and the possible 'bench-to-bedside' utility in patient diagnosis and care.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Miaoyun Qiu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Hannah Melchinger
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth E Gardiner
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
17
|
Turton HA, Pickworth J, Paterson GG, Lawrie A, Baillie JK, Thompson AAR. Soluble P-Selectin and von Willebrand Factor Rise in Healthy Volunteers Following Non-exertional Ascent to High Altitude. Front Physiol 2022; 13:825819. [PMID: 35250627 PMCID: PMC8889065 DOI: 10.3389/fphys.2022.825819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Reduced oxygen tensions experienced at high altitudes are thought to predispose to thrombosis, yet there are few studies linking hypoxia, platelet activation, and thrombosis. Reports of platelet phenotypes in hypoxia are inconsistent, perhaps due to differing degrees of hypoxia experienced and the duration of exposure. This study aimed to investigate the relationship between soluble P-selectin, a marker of platelet activation, and von Willebrand factor (vWF) on exposure to hypoxia. We measured plasma concentrations of P-selectin and vWF in sixteen healthy volunteers before, during and after the APEX 2 expedition. APEX 2 consisted of a non-exertional ascent to 5,200 m, followed by 7 consecutive days at high altitude. We showed that high altitude significantly increased mean plasma P-selectin and vWF compared to pre-expedition levels. Both plasma marker levels returned to baseline post-expedition. We found a strong positive correlation between vWF and P-selectin, but no association between P-selectin and platelet count. Our results are consistent with previous work showing evidence of platelet activation at high altitude and demonstrate that the rise in P-selectin is not simply due to an increase in platelet count. As vWF and P-selectin could be derived from either platelets or endothelial cells, further work assessing more specific markers of endothelial activation is proposed to provide insight into the source of these potential pro-thrombotic biomarkers at altitude.
Collapse
Affiliation(s)
- Helena A. Turton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Josephine Pickworth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Gordon G. Paterson
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Centre for Primary Care and Public Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - J. Kenneth Baillie
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - A. A. Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
- Apex (Altitude Physiology Expeditions, SC030345), Edinburgh, United Kingdom
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
18
|
Schrottmaier WC, Mussbacher M, Salzmann M, Kral-Pointner JB, Assinger A. PI3K Isoform Signalling in Platelets. Curr Top Microbiol Immunol 2022; 436:255-285. [PMID: 36243848 DOI: 10.1007/978-3-031-06566-8_11] [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] [Indexed: 06/16/2023]
Abstract
Platelets are unique anucleated blood cells that constantly patrol the vasculature to seal and prevent injuries in a process termed haemostasis. Thereby they rapidly adhere to the subendothelial matrix and recruit further platelets, resulting in platelet aggregates. Apart from their central role in haemostasis, they also kept some of their features inherited by their evolutionary ancestor-the haemocyte, which was also involved in immune defences. Together with leukocytes, platelets fight pathogenic invaders and guide many immune processes. In addition, they rely on several signalling pathways which are also relevant to immune cells. Among these, one of the central signalling hubs is the PI3K pathway. Signalling processes in platelets are unique as they lack a nucleus and therefore transcriptional regulation is absent. As a result, PI3K subclasses fulfil distinct roles in platelets compared to other cells. In contrast to leukocytes, the central PI3K subclass in platelet signalling is PI3K class Iβ, which underlines the uniqueness of this cell type and opens new ways for potential platelet-specific pharmacologic inhibition. An overview of platelet function and signalling with emphasis on PI3K subclasses and their respective inhibitors is given in this chapter.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marion Mussbacher
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA
| | - Manuel Salzmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Julia B Kral-Pointner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Platelet Function, Role in Thrombosis, Inflammation, and Consequences in Chronic Myeloproliferative Disorders. Cells 2021; 10:cells10113034. [PMID: 34831257 PMCID: PMC8616365 DOI: 10.3390/cells10113034] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 12/03/2022] Open
Abstract
Platelets are conventionally defined as playing a vital role in homeostasis and thrombosis. This role has over the years transformed as knowledge regarding platelets has expanded to include inflammation, cancer progression, and metastasis. Upon platelet activation and subsequent aggregation, platelets release a host of various factors, including numerous pro-inflammatory factors. These pro-inflammatory factors are recruiters and activators of leukocytes, aiding in platelets’ immune regulating function and inflammatory function. These various platelet functions are interrelated; activation of the inflammatory function results in thrombosis and, moreover, in various disease conditions, can result in worsened or chronic pathogenesis, including cancer. The role and contribution of platelets in a multitude of pathophysiological events during hemostasis, thrombosis, inflammation, cancer progression, and metastasis is an important focus for ongoing research. Platelet activation as discussed here is present in all platelet functionalities and can result in a multitude of factors and signaling pathways being activated. The cross-talk between inflammation, cancer, and platelets is therefore an ideal target for research and treatment strategies through antiplatelet therapy. Despite the knowledge implicating platelets in these mentioned processes, there is, nevertheless, limited literature available on the involvement and impact of platelets in many diseases, including myeloproliferative neoplasms. The extensive role platelets play in the processes discussed here is irrefutable, yet we do not fully understand the complete interrelation and extent of these processes.
Collapse
|
20
|
Ectodomain shedding by ADAM proteases as a central regulator in kidney physiology and disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119165. [PMID: 34699872 DOI: 10.1016/j.bbamcr.2021.119165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 11/20/2022]
Abstract
Besides its involvement in blood and bone physiology, the kidney's main function is to filter substances and thereby regulate the electrolyte composition of body fluids, acid-base balance and toxin removal. Depending on underlying conditions, the nephron must undergo remodeling and cellular adaptations. The proteolytic removal of cell surface proteins via ectodomain shedding by A Disintegrin and Metalloproteases (ADAMs) is of importance for the regulation of cell-cell and cell-matrix adhesion of renal cells. ADAM10 controls glomerular and tubule development in a Notch1 signaling-dependent manner and regulates brush border composition. ADAM17 regulates the renin angiotensin system and is together with ADAM10 involved in calcium phosphate homeostasis. In kidney disease ADAMs, especially ADAM17 contribute to inflammation through their involvement in IL-6 trans-signaling, Notch-, epithelial growth factor receptor-, and tumor necrosis factor α signaling. ADAMs are interesting drug targets to reduce the inflammatory burden, defective cell adhesion and impaired signaling pathways in kidney diseases.
Collapse
|
21
|
Abstract
Upon activation, platelets release a plethora of factors which help to mediate their dynamic functions in hemostasis, inflammation, wound healing, tumor metastasis and angiogenesis. The majority of these bioactive molecules are released from α-granules, which are unique to platelets, and contain an incredibly diverse repertoire of cargo including; integral membrane proteins, pro-coagulant molecules, chemokines, mitogenic, growth and angiogenic factors, adhesion proteins, and microbicidal proteins. Clinically, activation of circulating platelets has increasingly been associated with various disease states. Biomarkers indicating the level of platelet activation in patients can therefore be useful tools to evaluate risk factors to predict future complications and determine treatment strategies or evaluate antiplatelet therapy. The irreversible nature of α-granule secretion makes it ideally suited as a marker of platelet activation. This review outlines the release and contents of platelet α-granules, as well as the membrane bound, and soluble α-granule cargo proteins that can be used as biomarkers of platelet activation.
Collapse
Affiliation(s)
- Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| |
Collapse
|
22
|
Ebermeyer T, Cognasse F, Berthelot P, Mismetti P, Garraud O, Hamzeh-Cognasse H. Platelet Innate Immune Receptors and TLRs: A Double-Edged Sword. Int J Mol Sci 2021; 22:ijms22157894. [PMID: 34360659 PMCID: PMC8347377 DOI: 10.3390/ijms22157894] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Platelets are hematopoietic cells whose main function has for a long time been considered to be the maintenance of vascular integrity. They have an essential role in the hemostatic response, but they also have functional capabilities that go far beyond it. This review will provide an overview of platelet functions. Indeed, stress signals may induce platelet apoptosis through proapoptotis or hemostasis receptors, necrosis, and even autophagy. Platelets also interact with immune cells and modulate immune responses in terms of activation, maturation, recruitment and cytokine secretion. This review will also show that platelets, thanks to their wide range of innate immune receptors, and in particular toll-like receptors, and can be considered sentinels actively participating in the immuno-surveillance of the body. We will discuss the diversity of platelet responses following the engagement of these receptors as well as the signaling pathways involved. Finally, we will show that while platelets contribute significantly, via their TLRs, to immune response and inflammation, these receptors also participate in the pathophysiological processes associated with various pathogens and diseases, including cancer and atherosclerosis.
Collapse
Affiliation(s)
- Théo Ebermeyer
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Fabrice Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Etablissement Français du Sang Auvergne-Rhône-Alpes, 25 bd Pasteur, F-42100 Saint-Étienne, France
| | - Philippe Berthelot
- Team GIMAP, CIRI—Centre International de Recherche en Infectiologie, Université de Lyon, U1111, UMR5308, F-69007 Lyon, France;
- Infectious Diseases Department, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Patrick Mismetti
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Department of Vascular Medicine and Therapeutics, INNOVTE, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Olivier Garraud
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Hind Hamzeh-Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Correspondence:
| |
Collapse
|
23
|
Zhou Y, Heitmann JS, Kropp KN, Hinterleitner M, Koch A, Hartkopf AD, Salih HR, Hinterleitner C, Maurer S. Regulation of Platelet-Derived ADAM17: A Biomarker Approach for Breast Cancer? Diagnostics (Basel) 2021; 11:diagnostics11071188. [PMID: 34208863 PMCID: PMC8305148 DOI: 10.3390/diagnostics11071188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/14/2021] [Accepted: 06/26/2021] [Indexed: 12/22/2022] Open
Abstract
Tumor progression and metastasis are critically dependent on the tumor microenvironment. A disintegrin and metalloproteinase 17 (ADAM17) is associated with shedding of several substrates involved in tumor progression and known to be expressed by platelets of healthy donors and patients with solid tumors. Here, we report that platelet-derived ADAM17 (pADAM17) is regulated upon platelet activation of breast cancer patients, but not of healthy individuals. The observed downregulation of pADAM17 on platelets of cancer patients correlated with clinical parameters related to tumor progression including tumor stage and the occurrence of metastasis. Our data identify an association between platelet activation, modulation of platelet-derived ADAM17, and metastasis. In conclusion, we demonstrate that further development of pADAM17 as a liquid biomarker is warranted for monitoring disease progression in breast cancer.
Collapse
Affiliation(s)
- Yanjun Zhou
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Jonas S. Heitmann
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Korbinian N. Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, 55131 Mainz, Germany;
| | - Martina Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - André Koch
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72076 Tuebingen, Germany; (A.K.); (A.D.H.)
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72076 Tuebingen, Germany; (A.K.); (A.D.H.)
| | - Helmut R. Salih
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Clemens Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, 72076 Tuebingen, Germany
- Correspondence:
| | - Stefanie Maurer
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany; (Y.Z.); (J.S.H.); (M.H.); (H.R.S.); (S.M.)
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
24
|
Baidildinova G, Nagy M, Jurk K, Wild PS, Ten Cate H, van der Meijden PEJ. Soluble Platelet Release Factors as Biomarkers for Cardiovascular Disease. Front Cardiovasc Med 2021; 8:684920. [PMID: 34235190 PMCID: PMC8255615 DOI: 10.3389/fcvm.2021.684920] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Platelets are the main players in thrombotic diseases, where activated platelets not only mediate thrombus formation but also are involved in multiple interactions with vascular cells, inflammatory components, and the coagulation system. Although in vitro reactivity of platelets provides information on the function of circulating platelets, it is not a full reflection of the in vivo activation state, which may be relevant for thrombotic risk assessment in various disease conditions. Therefore, studying release markers of activated platelets in plasma is of interest. While this type of study has been done for decades, there are several new discoveries that highlight the need for a critical assessment of the available tests and indications for platelet release products. First, new insights have shown that platelets are not only prominent players in arterial vascular disease, but also in venous thromboembolism and atrial fibrillation. Second, knowledge of the platelet proteome has dramatically expanded over the past years, which contributed to an increasing array of tests for proteins released and shed from platelets upon activation. Identification of changes in the level of plasma biomarkers associated with upcoming thromboembolic events allows timely and individualized adjustment of the treatment strategy to prevent disease aggravation. Therefore, biomarkers of platelet activation may become a valuable instrument for acute event prognosis. In this narrative review based on a systematic search of the literature, we summarize the process of platelet activation and release products, discuss the clinical context in which platelet release products have been measured as well as the potential clinical relevance.
Collapse
Affiliation(s)
- Gaukhar Baidildinova
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Philipp S Wild
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hugo Ten Cate
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| | - Paola E J van der Meijden
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
25
|
Huang J, Swieringa F, Solari FA, Provenzale I, Grassi L, De Simone I, Baaten CCFMJ, Cavill R, Sickmann A, Frontini M, Heemskerk JWM. Assessment of a complete and classified platelet proteome from genome-wide transcripts of human platelets and megakaryocytes covering platelet functions. Sci Rep 2021; 11:12358. [PMID: 34117303 PMCID: PMC8196183 DOI: 10.1038/s41598-021-91661-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Novel platelet and megakaryocyte transcriptome analysis allows prediction of the full or theoretical proteome of a representative human platelet. Here, we integrated the established platelet proteomes from six cohorts of healthy subjects, encompassing 5.2 k proteins, with two novel genome-wide transcriptomes (57.8 k mRNAs). For 14.8 k protein-coding transcripts, we assigned the proteins to 21 UniProt-based classes, based on their preferential intracellular localization and presumed function. This classified transcriptome-proteome profile of platelets revealed: (i) Absence of 37.2 k genome-wide transcripts. (ii) High quantitative similarity of platelet and megakaryocyte transcriptomes (R = 0.75) for 14.8 k protein-coding genes, but not for 3.8 k RNA genes or 1.9 k pseudogenes (R = 0.43-0.54), suggesting redistribution of mRNAs upon platelet shedding from megakaryocytes. (iii) Copy numbers of 3.5 k proteins that were restricted in size by the corresponding transcript levels (iv) Near complete coverage of identified proteins in the relevant transcriptome (log2fpkm > 0.20) except for plasma-derived secretory proteins, pointing to adhesion and uptake of such proteins. (v) Underrepresentation in the identified proteome of nuclear-related, membrane and signaling proteins, as well proteins with low-level transcripts. We then constructed a prediction model, based on protein function, transcript level and (peri)nuclear localization, and calculated the achievable proteome at ~ 10 k proteins. Model validation identified 1.0 k additional proteins in the predicted classes. Network and database analysis revealed the presence of 2.4 k proteins with a possible role in thrombosis and hemostasis, and 138 proteins linked to platelet-related disorders. This genome-wide platelet transcriptome and (non)identified proteome database thus provides a scaffold for discovering the roles of unknown platelet proteins in health and disease.
Collapse
Affiliation(s)
- Jingnan Huang
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany.
| | - Frauke Swieringa
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
| | - Isabella Provenzale
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge, UK
| | - Ilaria De Simone
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Constance C F M J Baaten
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH, Aachen, Germany
| | - Rachel Cavill
- Department of Data Science and Knowledge Engineering, FSE, Maastricht University, Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge, UK
- Institute of Biomedical & Clinical Science, College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
26
|
The Missing Protein: Is T-Cadherin a Previously Unknown GPI-Anchored Receptor on Platelets? MEMBRANES 2021; 11:membranes11030218. [PMID: 33808741 PMCID: PMC8003554 DOI: 10.3390/membranes11030218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/01/2022]
Abstract
The membrane of platelets contains at least one uncharacterized glycosylphosphatidylinositol (GPI)-anchored protein according to the literature. Moreover, there is not enough knowledge on the receptor of low-density lipoproteins (LDL) mediating rapid Ca2+ signaling in platelets. Coincidentally, expression of a GPI-anchored protein T-cadherin increases LDL-induced Ca2+ signaling in nucleated cells. Here we showed evidence that supports the hypothesis about the presence of T-cadherin on platelets. The presence of T-cadherin on the surface of platelets and megakaryocytes was proven using antibodies whose specificity was tested on several negative and positive control cells by flow cytometry and confocal microscopy. Using phosphatidylinositol-specific phospholipase C, the presence of glycosylphosphatidylinositol anchor in the platelet T-cadherin form as well as in other known forms was confirmed. We showed by immunoblotting that the significant part of T-cadherin was detected in specific membrane domains (detergent Triton X-114 resistant) and the molecular weight of this newly identified protein was greater than that of T-cadherin from nucleated cells. Nevertheless, polymerase chain reaction data confirmed only the presence of isoform-1 of T-cadherin in platelets and megakaryocytes, which was also present in nucleated cells. We observed the redistribution of this newly identified protein after the activation of platelets, but only further work may explain its functional importance. Thus, our data described T-cadherin with some post-translational modifications as a new GPI-anchored protein on human platelets.
Collapse
|
27
|
Branfield S, Washington AV. The enigmatic nature of the triggering receptor expressed in myeloid cells -1 (TLT- 1). Platelets 2021; 32:753-760. [PMID: 33560928 DOI: 10.1080/09537104.2021.1881948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Receptors are important pharmacological targets on cells. The Triggering Receptor Expressed on Myeloid Cells (TREM) - Like Transcript - 1 is an abundant, yet little understood, platelet receptor. It is a single Ig domain containing receptor isolated in the α-granules of resting platelets and brought to the platelet surface upon activation. On platelets, the integrin αIIbβ3 is the major receptor having roughly 80,000 copies. αIIbβ3 is a heterodimeric multidomain structure that mediates platelet aggregation through its interaction with the plasma protein fibrinogen. Anti-platelet drugs have successfully targeted αIIbβ3 to control thrombosis. Like αIIbβ3, TLT-1 also binds fibrinogen, making its role in platelet function somewhat obscure. In this review, we highlight the known structural features of TLT-1 and present the challenges of understanding TLT-1 function. In our analysis of the dynamics of the platelet surface after activation we propose a model in which TLT-1 supports αIIbβ3 function as a mechanoreceptor that may direct platelets toward immune function.
Collapse
Affiliation(s)
- Siobhan Branfield
- , Department of Biology, University of Puerto Rico- Rio Piedras- Molecular Science Research Center, San Juan, Puerto Rico
| | - A Valance Washington
- , Department of Biology, University of Puerto Rico- Rio Piedras- Molecular Science Research Center, San Juan, Puerto Rico
| |
Collapse
|
28
|
Abstract
Cardiovascular pathologies are often induced by inflammation. The associated changes in the inflammatory response influence vascular endothelial biology; they complicate the extent of ischaemia and reperfusion injury, direct the migration of immune competent cells and activate platelets. The initiation and progression of inflammation is regulated by the classical paradigm through the system of cytokines and chemokines. Therapeutic approaches have previously used this knowledge to control the extent of cardiovascular changes with varying degrees of success. Neuronal guidance proteins (NGPs) have emerged in recent years and have been shown to be significantly involved in the control of tissue inflammation and the mechanisms of immune cell activation. Therefore, proteins of this class might be used in the future as targets to control the extent of inflammation in the cardiovascular system. In this review, we describe the role of NGPs during cardiovascular inflammation and highlight potential therapeutic options that could be explored in the future.
Collapse
|
29
|
Wu J, Heemskerk JWM, Baaten CCFMJ. Platelet Membrane Receptor Proteolysis: Implications for Platelet Function. Front Cardiovasc Med 2021; 7:608391. [PMID: 33490118 PMCID: PMC7820117 DOI: 10.3389/fcvm.2020.608391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
The activities of adhesion and signaling receptors in platelets are controlled by several mechanisms. An important way of regulation is provided by proteolytic cleavage of several of these receptors, leading to either a gain or a loss of platelet function. The proteases involved are of different origins and types: (i) present as precursor in plasma, (ii) secreted into the plasma by activated platelets or other blood cells, or (iii) intracellularly activated and cleaving cytosolic receptor domains. We provide a comprehensive overview of the proteases acting on the platelet membrane. We describe how these are activated, which are their target proteins, and how their proteolytic activity modulates platelet functions. The review focuses on coagulation-related proteases, plasmin, matrix metalloproteinases, ADAM(TS) isoforms, cathepsins, caspases, and calpains. We also describe how the proteolytic activities are determined by different platelet populations in a thrombus and conversely how proteolysis contributes to the formation of such populations.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| |
Collapse
|
30
|
Kyselova A, Elgheznawy A, Wittig I, Heidler J, Mann AW, Ruf W, Fleming I, Randriamboavonjy V. Platelet-derived calpain cleaves the endothelial protease-activated receptor 1 to induce vascular inflammation in diabetes. Basic Res Cardiol 2020; 115:75. [PMID: 33258989 PMCID: PMC7716944 DOI: 10.1007/s00395-020-00833-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus is a major risk factor for cardiovascular disease. Platelets from diabetic patients are hyperreactive and release microparticles that carry activated cysteine proteases or calpains. Whether platelet-derived calpains contribute to the development of vascular complications in diabetes is unknown. Here we report that platelet-derived calpain1 (CAPN1) cleaves the protease-activated receptor 1 (PAR-1) on the surface of endothelial cells, which then initiates a signaling cascade that includes the activation of the tumor necrosis factor (TNF)-α converting enzyme (TACE). The latter elicits the shedding of the endothelial protein C receptor and the generation of TNF-α, which in turn, induces intracellular adhesion molecule (ICAM)-1 expression to promote monocyte adhesion. All of the effects of CAPN1 were mimicked by platelet-derived microparticles from diabetic patients or from wild-type mice but not from CAPN1−/− mice, and were not observed in PAR-1-deficient endothelial cells. Importantly, aortae from diabetic mice expressed less PAR-1 but more ICAM-1 than non-diabetic mice, effects that were prevented by treating diabetic mice with a calpain inhibitor as well as by the platelet specific deletion of CAPN1. Thus, platelet-derived CAPN1 contributes to the initiation of the sterile vascular inflammation associated with diabetes via the cleavage of PAR-1 and the release of TNF-α from the endothelial cell surface.
Collapse
Affiliation(s)
- Anastasia Kyselova
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Ilka Wittig
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt am Main, Germany
| | - Juliana Heidler
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt am Main, Germany
| | | | - Wolfram Ruf
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
31
|
A disintegrin and metalloproteinase domain 17-epidermal growth factor receptor signaling contributes to oral cancer pain. Pain 2020; 161:2330-2343. [PMID: 32453136 PMCID: PMC9244849 DOI: 10.1097/j.pain.0000000000001926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer cells secrete pronociceptive mediators that sensitize adjacent sensory neurons and cause pain. Identification and characterization of these mediators could pinpoint novel targets for cancer pain treatment. In this study, we identified candidate genes in cancer cell lines that encode for secreted or cell surface proteins that may drive nociception. To undertake this work, we used an acute cancer pain mouse model, transcriptomic analysis of publicly available human tumor-derived cell line data, and a literature review. Cancer cell line supernatants were assigned a phenotype based on evoked nociceptive behavior in an acute cancer pain mouse model. We compared gene expression data from nociceptive and nonnociceptive cell lines. Our analyses revealed differentially expressed genes and pathways; many of the identified genes were not previously associated with cancer pain signaling. Epidermal growth factor receptor (EGFR) and disintegrin metalloprotease domain 17 (ADAM17) were identified as potential targets among the differentially expressed genes. We found that the nociceptive cell lines contained significantly more ADAM17 protein in the cell culture supernatant compared to nonnociceptive cell lines. Cytoplasmic EGFR was present in almost all (>90%) tongue primary afferent neurons in mice. Monoclonal antibody against EGFR, cetuximab, inhibited cell line supernatant-induced nociceptive behavior in an acute oral cancer pain mouse model. We infer from these data that ADAM17-EGFR signaling is involved in cancer mediator-induced nociception. The differentially expressed genes and their secreted protein products may serve as candidate therapeutic targets for oral cancer pain and warrant further evaluation.
Collapse
|
32
|
Schuhmann MK, Stoll G, Bieber M, Vögtle T, Hofmann S, Klaus V, Kraft P, Seyhan M, Kollikowski AM, Papp L, Heuschmann PU, Pham M, Nieswandt B, Stegner D. CD84 Links T Cell and Platelet Activity in Cerebral Thrombo-Inflammation in Acute Stroke. Circ Res 2020; 127:1023-1035. [PMID: 32762491 PMCID: PMC7508294 DOI: 10.1161/circresaha.120.316655] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 01/01/2023]
Abstract
RATIONALE Ischemic stroke is a leading cause of morbidity and mortality worldwide. Recanalization of the occluded vessel is essential but not sufficient to guarantee brain salvage. Experimental and clinical data suggest that infarcts often develop further due to a thromboinflammatory process critically involving platelets and T cells, but the underlying mechanisms are unknown. OBJECTIVE We aimed to determine the role of CD (cluster of differentiation)-84 in acute ischemic stroke after recanalization and to dissect the underlying molecular thromboinflammatory mechanisms. METHODS AND RESULTS Here, we show that mice lacking CD84-a homophilic immunoreceptor of the SLAM (signaling lymphocyte activation molecule) family-on either platelets or T cells displayed reduced cerebral CD4+ T-cell infiltration and thrombotic activity following experimental stroke resulting in reduced neurological damage. In vitro, platelet-derived soluble CD84 enhanced motility of wild-type but not of Cd84-/- CD4+ T cells suggesting homophilic CD84 interactions to drive this process. Clinically, human arterial blood directly sampled from the ischemic cerebral circulation indicated local shedding of platelet CD84. Moreover, high platelet CD84 expression levels were associated with poor outcome in patients with stroke. CONCLUSIONS These results establish CD84 as a critical pathogenic effector and thus a potential pharmacological target in ischemic stroke.
Collapse
Affiliation(s)
- Michael K. Schuhmann
- From the Department of Neurology (M.K.S., G.S., M.B., P.K., L.P.), University Hospital Würzburg, Germany
| | - Guido Stoll
- From the Department of Neurology (M.K.S., G.S., M.B., P.K., L.P.), University Hospital Würzburg, Germany
| | - Michael Bieber
- From the Department of Neurology (M.K.S., G.S., M.B., P.K., L.P.), University Hospital Würzburg, Germany
| | - Timo Vögtle
- Department I, Institute of Experimental Biomedicine (T.V., S.H., V.K., B.N., D.S.), University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging (T.V., S.H., B.N., D.S.), University of Würzburg, Germany
| | - Sebastian Hofmann
- Department I, Institute of Experimental Biomedicine (T.V., S.H., V.K., B.N., D.S.), University Hospital Würzburg, Germany
| | - Vanessa Klaus
- Department I, Institute of Experimental Biomedicine (T.V., S.H., V.K., B.N., D.S.), University Hospital Würzburg, Germany
| | - Peter Kraft
- From the Department of Neurology (M.K.S., G.S., M.B., P.K., L.P.), University Hospital Würzburg, Germany
- Department of Neurology, Klinikum Main-Spessart, Lohr, Germany (P.K.)
| | - Mert Seyhan
- Institute of Clinical Epidemiology and Biometry (M.S., P.U.H.), University of Würzburg, Germany
| | | | - Lena Papp
- From the Department of Neurology (M.K.S., G.S., M.B., P.K., L.P.), University Hospital Würzburg, Germany
| | - Peter U. Heuschmann
- Institute of Clinical Epidemiology and Biometry (M.S., P.U.H.), University of Würzburg, Germany
| | - Mirko Pham
- Department of Neuroradiology (A.M.K., M.P.), University Hospital Würzburg, Germany
| | - Bernhard Nieswandt
- Rudolf Virchow Center for Integrative and Translational Bioimaging (T.V., S.H., B.N., D.S.), University of Würzburg, Germany
| | - David Stegner
- Department I, Institute of Experimental Biomedicine (T.V., S.H., V.K., B.N., D.S.), University Hospital Würzburg, Germany
- Rudolf Virchow Center for Integrative and Translational Bioimaging (T.V., S.H., B.N., D.S.), University of Würzburg, Germany
| |
Collapse
|
33
|
Ibrutinib, but not zanubrutinib, induces platelet receptor shedding of GPIb-IX-V complex and integrin αIIbβ3 in mice and humans. Blood Adv 2020; 3:4298-4311. [PMID: 31869418 DOI: 10.1182/bloodadvances.2019000640] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/06/2019] [Indexed: 11/20/2022] Open
Abstract
The Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has proven to be efficacious in the treatment of B-cell chronic lymphocytic leukemia (B-CLL) and related diseases. However, a major adverse side effect of ibrutinib is bleeding, including major hemorrhages. The bleeding associated with ibrutinib use is thought to be due to a combination of on-target irreversible Btk inhibition, as well as off-target inhibition of other kinases, including EGFR, ITK, JAK3, and Tec kinase. In this study, we investigated the effects of ibrutinib vs zanubrutinib (a more selective Btk inhibitor) on platelet activation, glycoprotein expression, and thrombus formation. Ibrutinib, but not zanubrutinib, induced a time- and dose-dependent shedding of GPIb-IX complex and integrin αIIbβ3, but not of GPVI and GPV, from the platelet surface. The shedding of GPIbα and GPIX was blocked by GM6001 and TAPI-2, an ADAM17 inhibitor but not ADAM10 inhibitor. Ibrutinib but not zanubrutinib treatment of human platelets increased ADAM17 activation. Pretreatment of C57BL/6 mice with ibrutinib (10 mg/kg), but not zanubrutinib (10 mg/kg), inhibited ex vivo and in vivo thrombus growth over time. Platelets from ibrutinib-treated patients with CLL showed reduced GPIb-IX complex and integrin αIIbβ3 surface expression and reduced ex vivo thrombus formation under arterial flow, which was not observed in zanubrutinib-treated patients. In mice, ibrutinib, but not zanubrutinib, led to increased soluble GPIbα and soluble αIIb levels in plasma. These data demonstrate that ibrutinib induces shedding of GPIbα and GPIX by an ADAM17-dependent mechanism and integrin αIIbβ3 by an unknown sheddase, and this process occurs in vivo to regulate thrombus formation.
Collapse
|
34
|
Andrade SS, Faria AVDS, Girão MJBC, Fuhler GM, Peppelenbosch MP, Ferreira-Halder CV. Biotech-Educated Platelets: Beyond Tissue Regeneration 2.0. Int J Mol Sci 2020; 21:E6061. [PMID: 32842455 PMCID: PMC7503652 DOI: 10.3390/ijms21176061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 11/21/2022] Open
Abstract
The increasing discoveries regarding the biology and functions of platelets in the last decade undoubtedly show that these cells are one of the most biotechnological human cells. This review summarizes new advances in platelet biology, functions, and new concepts of biotech-educated platelets that connect advanced biomimetic science to platelet-based additive manufacturing for tissue regeneration. As highly responsive and secretory cells, platelets could be explored to develop solutions that alter injured microenvironments through platelet-based synthetic biomaterials with instructive extracellular cues for morphogenesis in tissue engineering beyond tissue regeneration 2.0.
Collapse
Affiliation(s)
| | - Alessandra Valéria de Sousa Faria
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil; (A.V.d.S.F.); (C.V.F.-H.)
- Department of Gastroenterology and Hepatology Medical Center Rotterdam, NL-3000 CA Rotterdam, The Netherlands; (G.M.F.); (M.P.P.)
| | | | - Gwenny M. Fuhler
- Department of Gastroenterology and Hepatology Medical Center Rotterdam, NL-3000 CA Rotterdam, The Netherlands; (G.M.F.); (M.P.P.)
| | - Maikel P. Peppelenbosch
- Department of Gastroenterology and Hepatology Medical Center Rotterdam, NL-3000 CA Rotterdam, The Netherlands; (G.M.F.); (M.P.P.)
| | - Carmen V. Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil; (A.V.d.S.F.); (C.V.F.-H.)
| |
Collapse
|
35
|
Iwagishi R, Tanaka R, Seto M, Takagi T, Norioka N, Ueyama T, Kawamura T, Takagi J, Ogawa Y, Shirakabe K. Negatively charged amino acids in the stalk region of membrane proteins reduce ectodomain shedding. J Biol Chem 2020; 295:12343-12352. [PMID: 32580944 DOI: 10.1074/jbc.ra120.013758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/11/2020] [Indexed: 01/12/2023] Open
Abstract
Ectodomain shedding is a post-translational modification mechanism by which the entire extracellular domain of membrane proteins is liberated through juxtamembrane processing. Because shedding rapidly and irreversibly alters the characteristics of cells, this process is properly regulated. However, the molecular mechanisms governing the propensity of membrane proteins to shedding are largely unknown. Here, we present evidence that negatively charged amino acids within the stalk region, an unstructured juxtamembrane region at which shedding occurs, contribute to shedding susceptibility. We show that two activated leukocyte cell adhesion molecule (ALCAM) protein variants produced by alternative splicing have different susceptibilities to ADAM metallopeptidase domain 17 (ADAM17)-mediated shedding. Of note, the inclusion of a stalk region encoded by a 39-bp-long alternative exon conferred shedding resistance. We found that this alternative exon encodes a large proportion of negatively charged amino acids, which we demonstrate are indispensable for conferring the shedding resistance. We also show that the introduction of negatively charged amino acids into the stalk region of shedding-susceptible ALCAM variant protein attenuates its shedding. Furthermore, we observed that negatively charged amino acids residing in the stalk region of Erb-B2 receptor tyrosine kinase 4 (ERBB4) are indispensable for its shedding resistance. Collectively, our results indicate that negatively charged amino acids within the stalk region interfere with the shedding of multiple membrane proteins. We conclude that the composition of the stalk region determines the shedding susceptibility of membrane proteins.
Collapse
Affiliation(s)
- Ryo Iwagishi
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Rika Tanaka
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Munenosuke Seto
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Tomoyo Takagi
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan
| | - Naoko Norioka
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Tomoe Ueyama
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan
| | - Teruhisa Kawamura
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan.,Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yoshihiro Ogawa
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medical and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Japan Agency for Medical Research and Development (AMED), Core Research for Evolutional Science and Technology (CREST), Tokyo, Japan
| | - Kyoko Shirakabe
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, Japan .,Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, Kusatsu, Japan.,Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
36
|
Maguire PB, Parsons ME, Szklanna PB, Zdanyte M, Münzer P, Chatterjee M, Wynne K, Rath D, Comer SP, Hayden M, Ní Áinle F, Gawaz M. Comparative Platelet Releasate Proteomic Profiling of Acute Coronary Syndrome versus Stable Coronary Artery Disease. Front Cardiovasc Med 2020; 7:101. [PMID: 32671099 PMCID: PMC7328343 DOI: 10.3389/fcvm.2020.00101] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/12/2020] [Indexed: 01/23/2023] Open
Abstract
Upon activation, platelets release a host of soluble and vesicular signals, collectively termed the “platelet releasate” (PR). The contents of this PR play a significant role in haemostasis, inflammation, and pathologic sequelae. Despite this, proteomic studies investigating the PR in coronary artery disease have not been performed. Here, we undertook a comparative label-free quantitative (LFQ) proteomic profiling of the 1 U/ml thrombin-induced PR from 13 acute coronary syndrome vs. 14 stable angina pectoris patients using a tandem mass spectrometry approach. Data are available via ProteomeXchange with identifier PXD009356. 318 PR proteins were identified across both cohorts with 9 proteins found to be differentially released, including tetranectin (CLEC3B), protein disulfide-isomerase-A3 (PDIA3), coagulation factor V (F5), and fibronectin (FN1). Strikingly, these 9 differential proteins were all associated with the gene ontology cellular component term “extracellular vesicle” and reduced levels of EVs were detected in the corresponding plasma of ST-segment elevation myocardial infarction (STEMI) patients. Network analysis revealed 3 proteins either reduced (F5; FN1) or absent (CLEC3B) in the PR of STEMI patients that are strongly connected to both the clotting cascade and major druggable targets on platelets. This moderated proteomic signature may prove useful for non-invasive risk assessment of the progression of coronary artery disease. These data further contribute to the growing evidence-base of using the platelet releasate as a predictor of pathological state and disease severity.
Collapse
Affiliation(s)
- Patricia B Maguire
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Institute for Discovery, University College Dublin, Dublin, Ireland.,Irish Centre for Vascular Biology, Dublin, Ireland
| | - Martin E Parsons
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Paulina B Szklanna
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,Irish Centre for Vascular Biology, Dublin, Ireland
| | - Monika Zdanyte
- Universitätsklinikum Tübingen, Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Tübingen, Germany
| | - Patrick Münzer
- Universitätsklinikum Tübingen, Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Tübingen, Germany
| | - Madhumita Chatterjee
- Universitätsklinikum Tübingen, Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Tübingen, Germany
| | - Kieran Wynne
- Proteomics Core, Conway Institute, University College Dublin, Dublin, Ireland
| | - Dominik Rath
- Universitätsklinikum Tübingen, Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Tübingen, Germany
| | - Shane P Comer
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Melanie Hayden
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Fionnuala Ní Áinle
- Conway SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland.,Irish Centre for Vascular Biology, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland.,Department of Haematology, Rotunda Hospital, Dublin, Ireland.,Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Meinrad Gawaz
- Universitätsklinikum Tübingen, Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Tübingen, Germany
| |
Collapse
|
37
|
Gutmann C, Joshi A, Mayr M. Platelet "-omics" in health and cardiovascular disease. Atherosclerosis 2020; 307:87-96. [PMID: 32646580 DOI: 10.1016/j.atherosclerosis.2020.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/28/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
The importance of platelets for cardiovascular disease was established as early as the 19th century. Their therapeutic inhibition stands alongside the biggest achievements in medicine. Still, certain aspects of platelet pathophysiology remain unclear. This includes platelet resistance to antiplatelet therapy and the contribution of platelets to vascular remodelling and extends beyond cardiovascular disease to haematological disorders and cancer. To address these gaps in our knowledge, a better understanding of the underlying molecular processes is needed. This will be enabled by technologies that capture dysregulated molecular processes and can integrate them into a broader network of biological systems. The advent of -omics technologies, such as mass spectrometry proteomics, metabolomics and lipidomics; highly multiplexed affinity-based proteomics; microarray- or RNA-sequencing-(RNA-seq)-based transcriptomics, and most recently ribosome footprint-based translatomics, has enabled a more holistic understanding of platelet biology. Most of these methods have already been applied to platelets, and this review will summarise this information and discuss future developments in this area of research.
Collapse
Affiliation(s)
- Clemens Gutmann
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Abhishek Joshi
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom.
| |
Collapse
|
38
|
Refaai MA, Conley GW, Hudson CA, Spinelli SL, Phipps RP, Morrell CN, Blumberg N, McRae HL. Evaluation of the procoagulant properties of a newly developed platelet modified lysate product. Transfusion 2020; 60:1579-1589. [PMID: 32415759 DOI: 10.1111/trf.15844] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Platelet transfusion is associated with logistical problems with the national storage guidelines of platelets. This results in decreased function in vivo as a result of the platelet storage lesion, and complications such as allergic or hemolytic reactions and thrombosis. We evaluated a new, freshly prepared platelet modified lysate (PML) product designed to be more procoagulant than fresh and stored platelets. METHODS Fresh platelets were concentrated, sonicated, and centrifuged to produce PML. Samples of both washed and unwashed PML were evaluated for particle size, concentration, and activity, and then tested for clot kinetics and thrombin generation. PML samples were also stored at various temperatures for durations up to 6 months and evaluated for clot kinetics and thrombin generation throughout. RESULTS PML showed significantly higher concentration of platelet microparticles, increased procoagulant properties, and increased thrombin generation as compared to fresh and stored platelets. In addition, PML maintained its clot kinetics over a 6-month storage period with variable storage conditions. CONCLUSIONS The newly proposed PML product is more procoagulant, stable, and has additional potential applications than currently available platelet products. Further studies will be performed to assess its functions in vivo and to assess thrombotic potential.
Collapse
Affiliation(s)
- Majed A Refaai
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Grace W Conley
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Sherry L Spinelli
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Richard P Phipps
- Department of Environmental Medicine and Microbiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Craig N Morrell
- Department of Cardiovascular Research, Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Neil Blumberg
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Hannah L McRae
- Transfusion Medicine Division, Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
39
|
Franco M, Reihani N, Dupuis L, Collec E, Billette de Villemeur T, Person M, Moussa F, Berger MG, Belmatoug N, Le Van Kim C. Semaphorin 7A: A novel marker of disease activity in Gaucher disease. Am J Hematol 2020; 95:483-491. [PMID: 31990411 DOI: 10.1002/ajh.25744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 01/13/2023]
Abstract
Gaucher disease (GD) is a recessively inherited lysosomal storage disorder in which sphingolipids accumulates in the macrophages that transform into Gaucher cells. A growing body of evidence indicates that red blood cells (RBCs) represent important actors in GD pathophysiology. We previously demonstrated that altered RBC properties including increased Lyso-GL1 levels, dyserythropoiesis, and iron metabolism defect in GD patients contribute to anemia and hyperferritinemia. Since RBC defects also correlated well with markers of GD severity and were normalized under enzyme replacement therapy (ERT), the identification of molecules that are deregulated in GD RBCs represents an important issue in the search of pertinent markers of the disease. Here, we found a decreased expression of the GPI-anchored cell surface protein Semaphorin 7A (Sema7A) in RBCs from untreated GD (GD UT) patients, in parallel with increased levels of the soluble form in the plasma. Sema7A plays a role in neural guidance, atherosclerosis, and inflammatory diseases and represents a promigratory cue in physiological and pathological conditions. We showed that the decreased expression of Sema7A in RBCs correlated with their abnormal properties and with markers of GD activity. Interestingly, ERT restored the level of Sema7A to normal values both in RBCs and in plasma from GD patients. We then proposed that SemaA7A represents a simple and pertinent marker of inflammation in GD. Finally, because Sema7A is known to regulate the activity of immune cells, the increased level of soluble Sema7A in GD patients could propagate inflammation in several tissues.
Collapse
Affiliation(s)
- Mélanie Franco
- Université de Paris, UMR_S1134, BIGR, Inserm, Institut National de Transfusion Sanguine, Laboratoire dʼExcellence GR‐Ex Paris France
| | - Nelly Reihani
- Université de Paris, UMR_S1134, BIGR, Inserm, Institut National de Transfusion Sanguine, Laboratoire dʼExcellence GR‐Ex Paris France
| | - Lucie Dupuis
- Université de Paris, UMR_S1134, BIGR, Inserm, Institut National de Transfusion Sanguine, Laboratoire dʼExcellence GR‐Ex Paris France
| | - Emmanuel Collec
- Université de Paris, UMR_S1134, BIGR, Inserm, Institut National de Transfusion Sanguine, Laboratoire dʼExcellence GR‐Ex Paris France
| | | | - Marine Person
- IUT Orsay, CNRS UMR 8000, Institut de Chimie Physique Orsay France
| | - Fathi Moussa
- IUT Orsay, CNRS UMR 8000, Institut de Chimie Physique Orsay France
| | - Marc G. Berger
- Université Clermont Auvergne, EA 7453 CHELTER Clermont‐Ferrand France
- CHU Clermont‐Ferrand, Service Hématologie Biologique, Hôpital Estaing Clermont‐Ferrand France
| | - Nadia Belmatoug
- Université de Paris, AP‐HP, CRML Maladies Lysosomales, Service de Médecine Interne, Hôpital Beaujon Clichy France
| | - Caroline Le Van Kim
- Université de Paris, UMR_S1134, BIGR, Inserm, Institut National de Transfusion Sanguine, Laboratoire dʼExcellence GR‐Ex Paris France
| |
Collapse
|
40
|
Schmoker AM, Perez Pearson LM, Cruz C, Colon Flores LG, Branfeild S, Pagán Torres FD, Fonseca K, Cantres YM, Salgado Ramirez CA, Melendez LM, Ballif BA, Washington AV. Defining the TLT-1 interactome from resting and activated human platelets. J Proteomics 2020; 215:103638. [PMID: 31923473 PMCID: PMC7044047 DOI: 10.1016/j.jprot.2020.103638] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022]
Abstract
The triggering receptor expressed on myeloid cells (TREM) protein family forms a class of type I transmembrane proteins expressed in immune cells that play important roles in innate and adaptive immune responses. The TREM family member TREM-like transcript 1 (TLT-1, also TREML1) is expressed in megakaryocytes and packaged into platelet granules. TLT-1 binds fibrinogen and plays a role in bleeding initiated by inflammatory insults. Here, we describe a proteomics screen that maps the TLT-1 interactome in resting and activated human platelets. Several identified TLT-1 interactors are involved in cell adhesion and migration, as well as platelet activation. Select interactors, including β3-integrin, RACK1, GRB2, and Rabs 5A, 7, and 11A, were additionally characterized in co-immunoprecipitation/immunoblotting experiments. Finally, several phosphorylation sites were found on immunoprecipitated TLT-1, including Thr280, a novel, regulated site on a conserved residue near the TLT-1 ITIM regulatory sequence. SIGNIFICANCE: Platelet function relies on the secretion of active molecules from intracellular vesicles, or granules, which contain soluble and membrane-bound proteins that are essential for platelet aggregation, coagulation reactions, and pathogen defense mechanisms. TLT-1 is sequestered in α-granules and transported to the plasma membrane, where it plays a unique role in hemostasis after inflammatory insults. Despite the known importance of TLT-1 in platelet biology, our knowledge of TLT-1 mechanistic signaling is limited. This study defines the TLT-1 interactome in resting and active human platelets, identifying several novel TLT-1 interactors, as well as TLT-1 phosphorylation sites, all with likely signaling implications in platelet aggregation dynamics.
Collapse
Affiliation(s)
- Anna M Schmoker
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA.
| | - Leishla M Perez Pearson
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA; Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Claudia Cruz
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA; Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Luis G Colon Flores
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Siobhan Branfeild
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Fabiola D Pagán Torres
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA
| | - Karmen Fonseca
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA
| | - Yadira M Cantres
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Carla A Salgado Ramirez
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Loyda M Melendez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA; Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA.
| | - A Valance Washington
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA.
| |
Collapse
|
41
|
Köhler D, Granja T, Volz J, Koeppen M, Langer HF, Hansmann G, Legchenko E, Geisler T, Bakchoul T, Eggstein C, Häberle HA, Nieswandt B, Rosenberger P. Red blood cell-derived semaphorin 7A promotes thrombo-inflammation in myocardial ischemia-reperfusion injury through platelet GPIb. Nat Commun 2020; 11:1315. [PMID: 32161256 PMCID: PMC7066172 DOI: 10.1038/s41467-020-14958-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 02/09/2020] [Indexed: 02/07/2023] Open
Abstract
Myocardial ischemia is one of the leading health problems worldwide. Therapy consists of the restitution of coronary perfusion which is followed by myocardial inflammation. Platelet–neutrophil interaction is a crucial process during inflammation, yet its consequences are not fully understood. Here, we show that platelet–neutrophil complexes (PNCs) are increased in patients with acute myocardial infarction and that this is associated with increased levels of neuronal guidance protein semaphorin 7A (SEMA7A). To investigate this further, we injected WT animals with Sema7a and found increased infarct size with increased numbers of PNCs. Experiments in genetically modified animals identify Sema7a on red blood cells to be crucial for this condition. Further studies revealed that Sema7a interacts with the platelet receptor glycoprotein Ib (GPIb). Treatment with anti-Sema7a antibody protected from myocardial tissue injury. In summary, we show that Sema7a binds to platelet GPIb and enhances platelet thrombo-inflammatory activity, aggravating post-ischemic myocardial tissue injury. Reperfusion injury following myocardial ischemia is aggravated by inflammation and platelet–neutrophil complex formation. Here the authors show that semaphorin 7A binds to platelet GPIb, enhancing platelet–neutrophil interaction and increasing post-ischemic myocardial tissue injury, and that blockage of semaphorin 7A is protective.
Collapse
Affiliation(s)
- David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Tiago Granja
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Julia Volz
- Institute of Experimental Biomedicine and Rudolf Virchow Center, Würzburg, Germany
| | - Michael Koeppen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Harald F Langer
- Department of Cardiology, University Hospital Lübeck, Lübeck, Germany
| | - Georg Hansmann
- Department of Pediatric Cardiology, Hannover Medical School, Lübeck, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology, Hannover Medical School, Lübeck, Germany
| | - Tobias Geisler
- Department of Cardiology, University Hospital, Tübingen, Germany
| | - Tamam Bakchoul
- Center for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Helene A Häberle
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine and Rudolf Virchow Center, Würzburg, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Tübingen, Germany.
| |
Collapse
|
42
|
Iragavarapu-Charyulu V, Wojcikiewicz E, Urdaneta A. Semaphorins in Angiogenesis and Autoimmune Diseases: Therapeutic Targets? Front Immunol 2020; 11:346. [PMID: 32210960 PMCID: PMC7066498 DOI: 10.3389/fimmu.2020.00346] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 02/12/2020] [Indexed: 01/17/2023] Open
Abstract
The axonal guidance molecules, semaphorins, have been described to function both physiologically and pathologically outside of the nervous system. In this review, we focus on the vertebrate semaphorins found in classes 3 through 7 and their roles in vascular development and autoimmune diseases. Recent studies indicate that while some of these vertebrate semaphorins promote angiogenesis, others have an angiostatic function. Since some semaphorins are also expressed by different immune cells and are known to modulate immune responses, they have been implicated in autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus and systemic sclerosis. We conclude this review by addressing strategies targeting semaphorins as potential therapeutic agents for angiogenesis and autoimmune diseases.
Collapse
Affiliation(s)
| | - Ewa Wojcikiewicz
- Department of Biomedical Sciences, Florida Atlantic University, Boca Raton, FL, United States
| | - Alexandra Urdaneta
- Department of Biomedical Sciences, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
43
|
Tarullo SE, Hill RC, Hansen KC, Behbod F, Borges VF, Nelson AC, Lyons TR. Postpartum breast cancer progression is driven by semaphorin 7a-mediated invasion and survival. Oncogene 2020; 39:2772-2785. [PMID: 32020054 PMCID: PMC7103487 DOI: 10.1038/s41388-020-1192-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 11/09/2022]
Abstract
Young women diagnosed with breast cancer (BC) have poor prognosis due to increased rates of metastasis. In addition, women diagnosed within 10 years of most recent childbirth are approximately three times more likely to develop metastasis than age- and stage-matched nulliparous women. We define these cases as postpartum BC (PPBC) and propose that the unique biology of the postpartum mammary gland drives tumor progression. Our published results revealed roles for SEMA7A in breast tumor cell growth, motility, invasion, and tumor-associated lymphangiogenesis, all of which are also increased in preclinical models of PPBC. However, whether SEMA7A drives progression in PPBC remains largely unexplored. Our results presented herein show that silencing of SEMA7A decreases tumor growth in a model of PPBC, while overexpression is sufficient to increase growth in nulliparous hosts. Further, we show that SEMA7A promotes multiple known drivers of PPBC progression including tumor-associated COX-2 expression and fibroblast-mediated collagen deposition in the tumor microenvironment. In addition, we show for the first time that SEMA7A-expressing cells deposit fibronectin to promote tumor cell survival. Finally, we show that co-expression of SEMA7A/COX-2/FN predicts for poor prognosis in breast cancer patient cohorts. These studies suggest SEMA7A as a key mediator of BC progression, and that targeting SEMA7A may open avenues for novel therapeutic strategies.
Collapse
Affiliation(s)
- Sarah E Tarullo
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ryan C Hill
- Department of Biochemistry and Molecular Genetics, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Fariba Behbod
- Division of Cancer and Developmental Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Cancer Center, Aurora, CO, 80045, USA
| | - Andrew C Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, CU Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Young Women's BC Translational Program, CU Anschutz Medical Campus, Aurora, CO, 80045, USA.
- University of Colorado Cancer Center, Aurora, CO, 80045, USA.
| |
Collapse
|
44
|
Maurer S, Kopp HG, Salih HR, Kropp KN. Modulation of Immune Responses by Platelet-Derived ADAM10. Front Immunol 2020; 11:44. [PMID: 32117229 PMCID: PMC7012935 DOI: 10.3389/fimmu.2020.00044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Platelets have a crucial function in maintaining hemostasis. However, beyond their role in coagulation and thrombus formation, platelets have been implicated to affect various pathophysiological conditions such as infectious diseases, autoimmune disorders, and cancer. It is well-established that platelets aid local cancer growth by providing growth factors or contributing to cancer angiogenesis. In addition, they promote metastasis, among others by facilitation of tumor cell-extravasation and epithelial-to-mesenchymal-like transition as well as protecting metastasizing cancer cells from immunosurveillance. A variety of membrane-bound and soluble platelet-derived factors are involved in these processes, and many aspects of platelet biology in both health and disease are regulated by platelet-associated metalloproteinases and their inhibitors. Platelets synthesize (i) members of the matrix metalloproteinase (MMP) family and also inhibitors of MMPs such as members of the "tissue inhibitor of metalloproteinases" (TIMP) family as well as (ii) members of the "a disintegrin and metalloproteinase" (ADAM) family including ADAM10. Notably, platelet-associated metalloproteinase activity not only influences functions of platelets themselves: platelets can also induce expression and/or release of metalloproteinases e.g., in leukocytes or cancer cells, and ADAMs are emerging as important components by which platelets directly affect other cell types and function. This review outlines the function of metalloproteinases in platelet biology with a focus on ADAM10 and discusses the role of platelet-derived metalloproteinases in the interaction of platelets with components of the immune system and/or cancer cells.
Collapse
Affiliation(s)
- Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hans-Georg Kopp
- Departments of Molecular Oncology and Thoracic Oncology, Robert-Bosch-Hospital Stuttgart, Stuttgart, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany
| | - Korbinian N Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, Mainz, Germany
| |
Collapse
|
45
|
Mathews JA, Ni YG, Wang C, Peterson JE, Ray C, Zhao X, Duan D, Hamon S, Allinson J, Hokom M, Wegner G. Considerations for Soluble Protein Biomarker Blood Sample Matrix Selection. AAPS JOURNAL 2020; 22:38. [PMID: 31997095 DOI: 10.1208/s12248-020-0412-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022]
Abstract
Blood-based soluble protein biomarkers provide invaluable clinical information about patients and are used as diagnostic, prognostic, and pharmacodynamic markers. The most commonly used blood sample matrices are serum and different types of plasma. In drug development research, the impact of sample matrix selection on successful protein biomarker quantification is sometimes overlooked. The sample matrix for a specific analyte is often chosen based on prior experience or literature searches, without good understanding of the possible effects on analyte quantification. Using a data set of 32 different soluble protein markers measured in matched serum and plasma samples, we examined the differences between serum and plasma and discussed how platelet or immune cell activation can change the quantified concentration of the analyte. We have also reviewed the effect of anticoagulant on analyte quantification. Finally, we provide specific recommendations for biomarker sample matrix selection and propose a systematic and data-driven approach for sample matrix selection. This review is intended to raise awareness of the impact and considerations of sample matrix selection on biomarker quantification.
Collapse
Affiliation(s)
- Joel A Mathews
- Ionis Pharmaceuticals, 2855 Gazelle Rd., Carlsbad, California, 92010, USA.
| | - Yan G Ni
- Regeneron, Tarrytown, New York, USA
| | | | | | - Chad Ray
- Zoetis, Kalamazoo, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Platelet heterogeneity in activation-induced glycoprotein shedding: functional effects. Blood Adv 2019; 2:2320-2331. [PMID: 30232085 DOI: 10.1182/bloodadvances.2017011544] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
The platelet receptors glycoprotein Ibα (GPIbα) and GPVI are known to be cleaved by members of a disintegrin and metalloprotease (ADAM) family (ADAM10 and ADAM17), but the mechanisms and consequences of this shedding are not well understood. Our results revealed that (1) glycoprotein shedding is confined to distinct platelet populations showing near-complete shedding, (2) the heterogeneity between (non)shed platelets is independent of agonist type but coincides with exposure of phosphatidylserine (PS), and (3) distinct pathways of shedding are induced by elevated Ca2+, low Ca2+ protein kinase C (PKC), or apoptotic activation. Furthermore, we found that receptor shedding reduces binding of von Willebrand factor, enhances binding of coagulation factors, and augments fibrin formation. In response to Ca2+-increasing agents, shedding of GPIbα was abolished by ADAM10/17 inhibition but not by blockage of calpain. Stimulation of PKC induced shedding of only GPIbα, which was annulled by kinase inhibition. The proapoptotic agent ABT-737 induced shedding, which was caspase dependent. In Scott syndrome platelets that are deficient in Ca2+-dependent PS exposure, shedding occurred normally, indicating that PS exposure is not a prerequisite for ADAM activity. In whole-blood thrombus formation, ADAM-dependent glycoprotein shedding enhanced thrombin generation and fibrin formation. Together, these findings indicate that 2 major activation pathways can evoke ADAM-mediated glycoprotein shedding in distinct platelet populations and that shedding modulates platelet function from less adhesive to more procoagulant.
Collapse
|
47
|
Utermöhlen O, Jakobshagen K, Blissenbach B, Wiegmann K, Merz T, Hefti JP, Krönke M. Emergence of AnnexinVpos CD31neg CD42blow/neg extracellular vesicles in plasma of humans at extreme altitude. PLoS One 2019; 14:e0220133. [PMID: 31369589 PMCID: PMC6675110 DOI: 10.1371/journal.pone.0220133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/09/2019] [Indexed: 12/02/2022] Open
Abstract
Background Hypobaric hypoxia has been reported to cause endothelial cell and platelet dysfunction implicated in the formation of microvascular lesions, and in its extremes may contribute to vascular leakage in high altitude pulmonary edema or blood brain barrier disruption leading to cerebral micro-hemorrhage (MH). Platelet function in the development of microvascular lesions remained ill defined, and is still incompletely understood. In this study platelet- and endothelial cell-derived extracellular vesicles (PEV and EEV, respectively) and cell adhesion molecules were characterized in plasma samples of members of a high altitude expedition to delineate the contribution of platelets and endothelial cells to hypobaric hypoxia-induced vascular dysfunction. Methods and findings In this observational study, platelet and endothelial cell-derived extracellular vesicles were analysed by flow-cytometry in plasma samples from 39 mountaineers participating in a medical research climbing expedition to Himlung Himal, Nepal, 7,050m asl. Megakaryocyte/platelet-derived AnnexinVpos, PECAM-1 (CD31) and glycoprotein-1b (GP1b, CD42b) positive extracellular vesicles (PEV) constituted the predominant fraction of EV in plasma samples up to 6,050m asl. Exposure to an altitude of 7,050m led to a marked decline of CD31pos CD42neg EEV as well as of CD31pos CD42bpos PEV at the same time giving rise to a quantitatively prevailing CD31neg CD42blow/neg subpopulation of AnnexinVpos EV. An almost hundredfold increase in the numbers of this previously unrecognized population of CD31neg CD42blow/neg EV was observed in all participants reaching 7,050m asl. Conclusions The emergence of CD31neg CD42blow/neg EV was observed in all participants and thus represents an early hypoxic marker at extreme altitude. Since CD31 and CD42b are required for platelet-endothelial cell interactions, these hypobaric hypoxia-dependent quantitative and phenotypic changes of AnnexinVpos EV subpopulations may serve as early and sensitive indicators of compromised vascular homeostasis.
Collapse
Affiliation(s)
- Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Kristin Jakobshagen
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
| | - Birgit Blissenbach
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Katja Wiegmann
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tobias Merz
- Department of Intensive Care Medicine, University Hospital and University of Bern, Bern, Switzerland
- Cardiovascular Intensive Care Unit, Auckland City Hospital, Auckland, New Zealand
| | - Jacqueline Pichler Hefti
- Department of Intensive Care Medicine, University Hospital and University of Bern, Bern, Switzerland
- Department of Pneumology, University Hospital and University of Bern, Bern, Switzerland
- * E-mail: (MK); (JP)
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- * E-mail: (MK); (JP)
| |
Collapse
|
48
|
Scanlon VM, Teixeira AM, Tyagi T, Zou S, Zhang PX, Booth CJ, Kowalska MA, Bao J, Hwa J, Hayes V, Marks MS, Poncz M, Krause DS. Epithelial (E)-Cadherin is a Novel Mediator of Platelet Aggregation and Clot Stability. Thromb Haemost 2019; 119:744-757. [PMID: 30861547 DOI: 10.1055/s-0039-1679908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cadherins play a major role in mediating cell-cell adhesion, which shares many parallels with platelet-platelet interactions during aggregate formation and clot stabilization. Platelets express epithelial (E)-cadherin, but its contribution to platelet function and/or platelet production is currently unknown. To assess the role of E-cadherin in platelet production and function in vitro and in vivo, we utilized a megakaryocyte-specific E-cadherin knockout mouse model. Loss of E-cadherin in megakaryocytes does not affect megakaryocyte maturation, platelet number or size. However, platelet dysfunction in the absence of E-cadherin is revealed when conditional knockout mice are challenged with acute antibody-mediated platelet depletion. Unlike wild-type mice that recover fully, knockout mice die within 72 hours post-antibody administration, likely from haemorrhage. Furthermore, conditional knockout mice have prolonged tail bleeding times, unstable clot formation, reduced clot retraction and reduced fibrin deposition in in vivo injury models. Murine platelet aggregation in vitro in response to thrombin and thrombin receptor activating peptide is compromised in E-cadherin null platelets, while aggregation in response to adenosine diphosphate (ADP) is not significantly different. Consistent with this, in vitro aggregation of primary human platelets in response to thrombin is decreased by an inhibitory E-cadherin antibody. Integrin activation and granule secretion in response to ADP and thrombin are not affected in E-cadherin null platelets, but Akt and glycogen synthase kinase 3β (GSK3β) activation are attenuated, suggesting a that E-cadherin contributes to aggregation, clot stabilization and retraction that is mediated by phosphoinositide 3-kinase/Akt/GSK3β signalling. In summary, E-cadherin plays a salient role in platelet aggregation and clot stability.
Collapse
Affiliation(s)
- Vanessa M Scanlon
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
| | | | - Tarun Tyagi
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
| | - Siying Zou
- Department of Cell Biology, Yale University, New Haven, Connecticut, United States
| | - Ping-Xia Zhang
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
| | - Carmen Jane Booth
- Department of Comparative Medicine, Yale University, New Haven, Connecticut, United States
| | - M Anna Kowalska
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Institute of Medical Biology, Polish Academy of Sciences, Philadelphia, Pennsylvania, United States
| | - Jialing Bao
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Department of Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - John Hwa
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
| | - Vincent Hayes
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Michael S Marks
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Department of Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Mortimer Poncz
- University of Pennsylvania, Philadelphia, Pennsylvania, United States.,Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States.,Department of Pathology, Yale University, New Haven, Connecticut, United States.,Department of Cell Biology, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
49
|
Jaiswal R, Sedger LM. Intercellular Vesicular Transfer by Exosomes, Microparticles and Oncosomes - Implications for Cancer Biology and Treatments. Front Oncol 2019; 9:125. [PMID: 30895170 PMCID: PMC6414436 DOI: 10.3389/fonc.2019.00125] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Intercellular communication is a normal feature of most physiological interactions between cells in healthy organisms. While cells communicate directly through intimate physiology contact, other mechanisms of communication exist, such as through the influence of soluble mediators such as growth factors, cytokines and chemokines. There is, however, yet another mechanism of intercellular communication that permits the exchange of information between cells through extracellular vesicles (EVs). EVs are microscopic (50 nm−10 μM) phospholipid bilayer enclosed entities produced by virtually all eukaryotic cells. EVs are abundant in the intracellular space and are present at a cells' normal microenvironment. Irrespective of the EV “donor” cell type, or the mechanism of EV biogenesis and production, or the size and EV composition, cancer cells have the potential to utilize EVs in a manner that enhances their survival. For example, cancer cell EV overproduction confers benefits to tumor growth, and tumor metastasis, compared with neighboring healthy cells. Herein, we summarize the current status of knowledge on different populations of EVs. We review the situations that regulate EV release, and the factors that instruct differential packaging or sorting of EV content. We then highlight the functions of cancer-cell derived EVs as they impact on cancer outcomes, promoting tumor progression, metastases, and the mechanisms by which they facilitate the creation of a pre-metastatic niche. The review finishes by focusing on the beneficial (and challenging) features of tumor-derived EVs that can be adapted and utilized for cancer treatments, including those already being investigated in human clinical trials.
Collapse
Affiliation(s)
- Ritu Jaiswal
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Lisa M Sedger
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
50
|
Differential protein expression of blood platelet components associated with adverse transfusion reactions. J Proteomics 2019; 194:25-36. [DOI: 10.1016/j.jprot.2018.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023]
|