1
|
Goubran H, Ahmed S, Ragab G, Seghatchian J, Burnouf T. Platelet proteomics: Clinical implications - Decoding the black box! Transfus Apher Sci 2025; 64:104060. [PMID: 39719751 DOI: 10.1016/j.transci.2024.104060] [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] [Indexed: 12/26/2024]
Abstract
Platelets are anucleate blood cells traditionally associated with hemostasis but now increasingly recognized for their multifaceted roles in immunity, inflammation, and tissue repair. Advances in platelet proteomics, employing high-throughput techniques such as mass spectrometry, have significantly enhanced our understanding of platelet biology and its clinical implications in transfusion medicine. Platelet proteomics offers a retrospective view of physiological and pathological changes over the platelet's 7-10-day lifespan, making it a unique tool for studying cumulative biological events. Recent applications include the identification of biomarkers for cardiovascular, infectious, autoimmune diseases and cancer. In neurodegeneration and aging, platelets have been explored for their shared molecular pathways with neurons, with findings implicating Tau, amyloid-beta, and alpha-synuclein as potential biomarkers. Proteomics is also emerging as an important factor in the development of evidence-based, tailor-made platelet-derived therapies. While promising, platelet proteomics requires further standardization and computational advances to support transitioning from research to routine clinical practice.
Collapse
Affiliation(s)
- Hadi Goubran
- Saskatoon Cancer Centre, Saskatoon, SK, Canada; Department of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Shahid Ahmed
- Saskatoon Cancer Centre, Saskatoon, SK, Canada; Department of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Gaafar Ragab
- Rheumatology and Immunology Unit, Internal Medicine Department, Cairo University, Egypt
| | - Jerard Seghatchian
- International Consultancy in Modern Personalized Blood Components Therapies and Innovative DDR Strategies, London, England, UK
| | - Thierry Burnouf
- Graduate Institute of Biological Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Gallagher LT, LaCroix I, Fields AT, Mitra S, Argabright A, D'Alessandro A, Erickson C, Nunez-Garcia B, Herrera-Rodriguez K, Chou YC, Stocker BW, Ramser BJ, Thielen O, Hallas W, Silliman CC, Kornblith LZ, Cohen MJ. Platelet releasates mitigate the endotheliopathy of trauma. J Trauma Acute Care Surg 2024; 97:738-746. [PMID: 38764145 PMCID: PMC11502277 DOI: 10.1097/ta.0000000000004342] [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: 05/21/2024]
Abstract
BACKGROUND Platelets are well known for their roles in hemostasis, but they also play a key role in thromboinflammatory pathways by regulating endothelial health, stimulating angiogenesis, and mediating host defense through both contact dependent and independent signaling. When activated, platelets degranulate releasing multiple active substances. We hypothesized that the soluble environment formed by trauma platelet releasates (TPR) attenuates thromboinflammation via mitigation of trauma induced endothelial permeability and metabolomic reprogramming. METHODS Blood was collected from injured and healthy patients to generate platelet releasates and plasma in parallel. Permeability of endothelial cells when exposed to TPR and plasma (TP) was assessed via resistance measurement by electric cell-substrate impedance sensing (ECIS). Endothelial cells treated with TPR and TP were subjected to mass spectrometry-based metabolomics. RESULTS TP increased endothelial permeability, whereas TPR decreased endothelial permeability when compared with untreated cells. When TP and TPR were mixed ex vivo, TPR mitigated TP-induced permeability, with significant increase in AUC compared with TP alone. Metabolomics of TPR and TP demonstrated disrupted redox reactions and anti-inflammatory mechanisms. CONCLUSION Trauma platelet releasates provide endothelial barrier protection against TP-induced endothelial permeability. Our findings highlight a potential beneficial action of activated platelets on the endothelium in injured patients through disrupted redox reactions and increased antioxidants. Our findings support that soluble signaling from platelet degranulation may mitigate the endotheliopathy of trauma. The clinical implications of this are that activated platelets may prove a promising therapeutic target in the complex integration of thrombosis, endotheliopathy, and inflammation in trauma.
Collapse
Affiliation(s)
- Lauren T Gallagher
- From the Department of Surgery (L.T.G., S.M., B.W.S., B.J.R., O.T., W.H., M.J.C.), Department of Biochemistry and Molecular Genetics (I.L.C., C.E.), University of Colorado, School of Medicine, Aurora, Colorado; Department of Surgery (A.T.F., B.N.-G., K.H.-R., Y.C.C., L.Z.K.), University of California, San Francisco, San Francisco, California; Mass Spectrometry Core Facility (A.A.), University of Colorado, School of Medicine; Department of Biochemistry and Molecular Genetics (A.D'A.), University of Colorado Anschutz Medical Campus, School of Medicine; Vitalant Research Institute, Department of Surgery (C.C.S.), Department of Pediatrics (C.C.S.), University of Colorado, School of Medicine, Aurora, Colorado; and Department of Laboratory Medicine (L.Z.K.), University of California, San Francisco, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Saba L, Maindarkar M, Johri AM, Mantella L, Laird JR, Khanna NN, Paraskevas KI, Ruzsa Z, Kalra MK, Fernandes JFE, Chaturvedi S, Nicolaides A, Rathore V, Singh N, Isenovic ER, Viswanathan V, Fouda MM, Suri JS. UltraAIGenomics: Artificial Intelligence-Based Cardiovascular Disease Risk Assessment by Fusion of Ultrasound-Based Radiomics and Genomics Features for Preventive, Personalized and Precision Medicine: A Narrative Review. Rev Cardiovasc Med 2024; 25:184. [PMID: 39076491 PMCID: PMC11267214 DOI: 10.31083/j.rcm2505184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/24/2024] [Accepted: 03/05/2024] [Indexed: 07/31/2024] Open
Abstract
Cardiovascular disease (CVD) diagnosis and treatment are challenging since symptoms appear late in the disease's progression. Despite clinical risk scores, cardiac event prediction is inadequate, and many at-risk patients are not adequately categorised by conventional risk factors alone. Integrating genomic-based biomarkers (GBBM), specifically those found in plasma and/or serum samples, along with novel non-invasive radiomic-based biomarkers (RBBM) such as plaque area and plaque burden can improve the overall specificity of CVD risk. This review proposes two hypotheses: (i) RBBM and GBBM biomarkers have a strong correlation and can be used to detect the severity of CVD and stroke precisely, and (ii) introduces a proposed artificial intelligence (AI)-based preventive, precision, and personalized ( aiP 3 ) CVD/Stroke risk model. The PRISMA search selected 246 studies for the CVD/Stroke risk. It showed that using the RBBM and GBBM biomarkers, deep learning (DL) modelscould be used for CVD/Stroke risk stratification in the aiP 3 framework. Furthermore, we present a concise overview of platelet function, complete blood count (CBC), and diagnostic methods. As part of the AI paradigm, we discuss explainability, pruning, bias, and benchmarking against previous studies and their potential impacts. The review proposes the integration of RBBM and GBBM, an innovative solution streamlined in the DL paradigm for predicting CVD/Stroke risk in the aiP 3 framework. The combination of RBBM and GBBM introduces a powerful CVD/Stroke risk assessment paradigm. aiP 3 model signifies a promising advancement in CVD/Stroke risk assessment.
Collapse
Affiliation(s)
- Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy
| | - Mahesh Maindarkar
- School of Bioengineering Sciences and Research, MIT Art, Design and Technology University, 412021 Pune, India
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
| | - Amer M. Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Laura Mantella
- Department of Medicine, Division of Cardiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St Helena, CA 94574, USA
| | - Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, 110001 New Delhi, India
| | | | - Zoltan Ruzsa
- Invasive Cardiology Division, University of Szeged, 6720 Szeged, Hungary
| | - Manudeep K. Kalra
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Seemant Chaturvedi
- Department of Neurology & Stroke Program, University of Maryland, Baltimore, MD 20742, USA
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre and University of Nicosia Medical School, 2368 Agios Dometios, Cyprus
| | - Vijay Rathore
- Nephrology Department, Kaiser Permanente, Sacramento, CA 95823, USA
| | - Narpinder Singh
- Department of Food Science and Technology, Graphic Era Deemed to be University, Dehradun, 248002 Uttarakhand, India
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, National Institute of The Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA
| | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA
- Department of Computer Engineering, Graphic Era Deemed to be University, Dehradun, 248002 Uttarakhand, India
| |
Collapse
|
4
|
Berry CE, Abbas DB, Lintel HA, Churukian AA, Griffin M, Guo JL, Cotterell AC, Parker JBL, Downer MA, Longaker MT, Wan DC. Adipose-Derived Stromal Cell-Based Therapies for Radiation-Induced Fibrosis. Adv Wound Care (New Rochelle) 2024; 13:235-252. [PMID: 36345216 PMCID: PMC11304913 DOI: 10.1089/wound.2022.0103] [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: 08/01/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Significance: Half of all cancer patients receive radiation therapy as a component of their treatment regimen, and the most common resulting complication is radiation-induced fibrosis (RIF) of the skin and soft tissue. This thickening of the dermis paired with decreased vascularity results in functional limitations and esthetic concerns and poses unique challenges when considering surgical exploration or reconstruction. Existing therapeutic options for RIF of the skin are limited both in scope and efficacy. Cell-based therapies have emerged as a promising means of utilizing regenerative cell populations to improve both functional and esthetic outcomes, and even as prophylaxis for RIF. Recent Advances: As one of the leading areas of cell-based therapy research, adipose-derived stromal cells (ADSCs) demonstrate significant therapeutic potential in the treatment of RIF. The introduction of the ADSC-augmented fat graft has shown clinical utility. Recent research dedicated to characterizing specific ADSC subpopulations points toward further granularity in understanding of the mechanisms driving the well-established clinical outcomes seen with fat grafting therapy. Critical Issues: Various animal models of RIF demonstrated improved clinical outcomes following treatment with cell-based therapies, but the cellular and molecular basis underlying these effects remains poorly understood. Future Directions: Recent literature has focused on improving the efficacy of cell-based therapies, most notably through (1) augmentation of fat grafts with platelet-rich plasma and (2) the modification of expressed RNA through epitranscriptomics. For the latter, new and promising gene targets continue to be identified which have the potential to reverse the effects of fibrosis by increasing angiogenesis, decreasing inflammation, and promoting adipogenesis.
Collapse
Affiliation(s)
- Charlotte E. Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Darren B. Abbas
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hendrik A. Lintel
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Andrew A. Churukian
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jason L. Guo
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Asha C. Cotterell
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jennifer B. Laufey Parker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Mauricio A. Downer
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
5
|
Russo I, Brookles CG, Barale C, Melchionda E, Mousavi AH, Biolè C, Chinaglia A, Bianco M. Current Strategies to Guide the Antiplatelet Therapy in Acute Coronary Syndromes. Int J Mol Sci 2024; 25:3981. [PMID: 38612792 PMCID: PMC11011739 DOI: 10.3390/ijms25073981] [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: 02/18/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
The role of antiplatelet therapy in patients with acute coronary syndromes is a moving target with considerable novelty in the last few years. The pathophysiological basis of the treatment depends on platelet biology and physiology, and the interplay between these aspects and clinical practice must guide the physician in determining the best therapeutic options for patients with acute coronary syndromes. In the present narrative review, we discuss the latest novelties in the antiplatelet therapy of patients with acute coronary syndromes. We start with a description of platelet biology and the role of the main platelet signal pathways involved in platelet aggregation during an acute coronary syndrome. Then, we present the latest evidence on the evaluation of platelet function, focusing on the strengths and weaknesses of each platelet's function test. We continue our review by describing the role of aspirin and P2Y12 inhibitors in the treatment of acute coronary syndromes, critically appraising the available evidence from clinical trials, and providing current international guidelines and recommendations. Finally, we describe alternative therapeutic regimens to standard dual antiplatelet therapy, in particular for patients at high bleeding risk. The aim of our review is to give a comprehensive representation of current data on antiplatelet therapy in patients with acute coronary syndromes that could be useful both for clinicians and basic science researchers to be up-to-date on this complex topic.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Turin, Italy; (I.R.); (C.B.); (E.M.)
| | - Carola Griffith Brookles
- Cardiology Division, San Luigi Gonzaga University Hospital, I-10043 Orbassano, Italy; (C.G.B.); (A.H.M.); (C.B.); (A.C.)
- Department of Medical Sciences, University of Turin, I-10124 Turin, Italy
| | - Cristina Barale
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Turin, Italy; (I.R.); (C.B.); (E.M.)
| | - Elena Melchionda
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Turin, Italy; (I.R.); (C.B.); (E.M.)
| | - Amir Hassan Mousavi
- Cardiology Division, San Luigi Gonzaga University Hospital, I-10043 Orbassano, Italy; (C.G.B.); (A.H.M.); (C.B.); (A.C.)
- Department of Medical Sciences, University of Turin, I-10124 Turin, Italy
| | - Carloalberto Biolè
- Cardiology Division, San Luigi Gonzaga University Hospital, I-10043 Orbassano, Italy; (C.G.B.); (A.H.M.); (C.B.); (A.C.)
| | - Alessandra Chinaglia
- Cardiology Division, San Luigi Gonzaga University Hospital, I-10043 Orbassano, Italy; (C.G.B.); (A.H.M.); (C.B.); (A.C.)
| | - Matteo Bianco
- Cardiology Division, San Luigi Gonzaga University Hospital, I-10043 Orbassano, Italy; (C.G.B.); (A.H.M.); (C.B.); (A.C.)
| |
Collapse
|
6
|
Franco R, Cervino G, Vazzana G, Rocca FD, Ferrari G, Cicciù M, Minervini G. Use of Concentrated Growth Factor (CGF) in Prosthetic-Guided Reconstruction on Two-Wall Bone Defect after Cystectomy: An Alternative to Traditional Regeneration. Eur J Dent 2024; 18:392-396. [PMID: 37311550 PMCID: PMC10959619 DOI: 10.1055/s-0043-1768973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Abstract
This clinical case report's objective was to describe an alternative technique executed to ensure bone regeneration after removing a cystic lesion in the upper jaw. The bone defect after the cystectomy was filled with autologous fibrin-rich clots containing concentrated growth factor (CGF). A 45-year-old female patient was presumed to have a cystic lesion with massive bone destruction on the vestibular and palatal walls between teeth 2.2 and 2.3. CGF was applied to fill the gap to promote the development of the bone. The tooth was asymptomatic and repair was still increasing steadily after a year, according to the results of the clinical and radiological follow-up assessment. This article describes a different way to treat a two-wall defect involving both the palatal and buccal bone, after removing a cystic lesion, with the use of CGF as an equivalent to the traditional use of autologous or heterologous bone. A promising substance for bone repair is CGF fibrin, which may encourage the growth of new bone in jaw deformities and promote bone tissue healing.
Collapse
Affiliation(s)
- Rocco Franco
- Department of Biomedicine and Prevention, University of University of Rome “Tor Vergata,” Rome, Italy
| | - Gabriele Cervino
- School of Dentistry Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | | | | | | | - Marco Cicciù
- School of Dentistry Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Giuseppe Minervini
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli,” Naples, Italy
| |
Collapse
|
7
|
Andersson LI, Sjöström DJ, Quach HQ, Hägerström K, Hurler L, Kajdácsi E, Cervenak L, Prohászka Z, Toonen EJM, Mohlin C, Mollnes TE, Sandgren P, Tjernberg I, Nilsson PH. Storage of Transfusion Platelet Concentrates Is Associated with Complement Activation and Reduced Ability of Platelets to Respond to Protease-Activated Receptor-1 and Thromboxane A2 Receptor. Int J Mol Sci 2024; 25:1091. [PMID: 38256162 PMCID: PMC10816124 DOI: 10.3390/ijms25021091] [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: 12/18/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Platelet activation and the complement system are mutually dependent. Here, we investigated the effects of storage time on complement activation and platelet function in routinely produced platelet concentrates. The platelet concentrates (n = 10) were stored at 22 °C for seven days and assessed daily for complement and platelet activation markers. Additionally, platelet function was analyzed in terms of their responsiveness to protease-activated receptor-1 (PAR-1) and thromboxane A2 receptor (TXA2R) activation and their capacity to adhere to collagen. Complement activation increased over the storage period for all analyzed markers, including the C1rs/C1-INH complex (fold change (FC) = 1.9; p < 0.001), MASP-1/C1-INH complex (FC = 2.0; p < 0.001), C4c (FC = 1.8, p < 0.001), C3bc (FC = 4.0; p < 0.01), and soluble C5b-9 (FC = 1.7, p < 0.001). Furthermore, the levels of soluble platelet activation markers increased in the concentrates over the seven-day period, including neutrophil-activating peptide-2 (FC = 2.5; p < 0.0001), transforming growth factor beta 1 (FC = 1.9; p < 0.001) and platelet factor 4 (FC = 2.1; p < 0.0001). The ability of platelets to respond to activation, as measured by surface expression of CD62P and CD63, decreased by 19% and 24% (p < 0.05) for PAR-1 and 69-72% (p < 0.05) for TXA2R activation, respectively, on Day 7 compared to Day 1. The extent of platelet binding to collagen was not significantly impaired during storage. In conclusion, we demonstrated that complement activation increased during the storage of platelets, and this correlated with increased platelet activation and a reduced ability of the platelets to respond to, primarily, TXA2R activation.
Collapse
Affiliation(s)
- Linnea I. Andersson
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Dick J. Sjöström
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA;
| | - Kim Hägerström
- Department of Clinical Chemistry and Transfusion Medicine, Region Kalmar County, 391 85 Kalmar, Sweden; (K.H.); (I.T.)
| | - Lisa Hurler
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary; (L.H.); (E.K.); (L.C.); (Z.P.)
| | | | - Camilla Mohlin
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, University of Oslo, 0372 Oslo, Norway;
- Research Laboratory, Nordland Hospital, 8005 Bodo, Norway
| | - Per Sandgren
- Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 171 77 Huddinge, Sweden;
| | - Ivar Tjernberg
- Department of Clinical Chemistry and Transfusion Medicine, Region Kalmar County, 391 85 Kalmar, Sweden; (K.H.); (I.T.)
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, 581 83 Linköping, Sweden
| | - Per H. Nilsson
- Department of Chemistry and Biomedicine, Linnaeus University, 391 82 Kalmar, Sweden; (L.I.A.); (D.J.S.); (C.M.)
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, 391 82 Kalmar, Sweden
| |
Collapse
|
8
|
Chen X, Mohammed AF, Li C. Assessment of the Clinical Value of Platelet-to-Lymphocyte Ratio in Patients with Hepatocellular Carcinoma. Clin Appl Thromb Hemost 2024; 30:10760296231221535. [PMID: 38591958 PMCID: PMC11005495 DOI: 10.1177/10760296231221535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 04/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with higher mortality as a result of poor prognosis and unavailability of effective treatment options. This study retrospectively analyzed the clinical value of platelet-to-lymphocyte ratio (PLR) to aid in differentiating early hepatocellular carcinoma from liver cirrhosis patients. Three hundred and nine (309) patients including 155 patients with hepatocellular carcinoma (HCC) and 154 patients with liver cirrhosis were enrolled in this study. General clinical characteristics and blood parameters of each patient were collected, calculated, and retrospectively analyzed. Mann-Whitney U test was calculated to compare the two groups. Receiver operating characteristics (ROC) curve was performed to investigate the diagnostic potential of PLR in the prediction of HCC at a cut-off with high accuracy (area under the curve [AUC]) > 0.80. Hemoglobin (HB) concentration, red blood cell (RBC) count, neutrophil (NEU) count, platelet count, platelet-to-lymphocyte ratio (PLR), and neutrophil-to-lymphocyte ratio (NLR) were significantly higher in the HCC patients than in the liver cirrhosis patients (p < 0.05). ROC curve analysis showed that the AUC, optimal cut-off value, sensitivity, and specificity of PLR to predict HCC patients were 0.912, 98.7, 81.2%, and 80.6% respectively. The results suggest that PLR is a potential biomarker that can be used to predict early HCC.
Collapse
Affiliation(s)
- Xu Chen
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Abdul Fatawu Mohammed
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Chengbin Li
- Department of Laboratory Medicine, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| |
Collapse
|
9
|
Tian J, Li XJ, Ma Y, Mai Z, Yang Y, Luo M, Xu W, Chen K, Chen X, Tang J, Cheng B, Cui X. Correlation of bioactive components of platelet rich plasma derived from human female adult peripheral blood and umbilical cord blood with age. Sci Rep 2023; 13:18428. [PMID: 37891219 PMCID: PMC10611812 DOI: 10.1038/s41598-023-45747-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023] Open
Abstract
Platelet-rich plasma (PRP) has gained significant attention in the field of regenerative medicine due to its potential therapeutic applications. However, few studies have reported the components, especially anti-ageing-related components, of PRP derived from umbilical cord blood (UCB). It is essential to understand the influence of age on the composition and efficacy of PRP to optimize its clinical use. The present study compared the concentrations of bioactive components in PRP from healthy female adults and UCB-derived PRP. PRP was obtained from blood samples from females in four age groups (12 per group): neonates (UCB donors) and adults aged 18-25, 26-45, and 46-65 years, respectively. The concentrations of epidermal growth factor, basic fibroblast growth factor-2 (FGF-2), insulin-like growth factor-1, platelet-derived growth factor-AA (PDGF-AA), PDGF-AB/BB, vascular endothelial growth factor A, RANTES, TIMP-1, TIMP-2, GDF11, and clusterin and activity of superoxide dismutase, catalase, and glutathione peroxidase (GPx) in the PRP samples were determined and compared among groups. Pairwise comparisons between the groups showed statistically significant differences in the concentrations of some bioactive components of PRP, such as FGF-2, PDGF-AB/BB, and clusterin, and GPx activity. UCB-derived PRP contains various active ingredients such as VEGF-A, CAT activity, and TIMP-2. Contrary to expectations, UCB-derived PRP did not show higher concentrations of the anti-ageing protein GDF11. Because UCB is a rich source of bioactive components with low immunogenicity, its use in PRP preparation is an important research direction for future studies.
Collapse
Affiliation(s)
- Ju Tian
- Department of Plastic Surgery, People's Hospital of Zhongshan City, Zhongshan, 528421, Guangdong, China
| | - Xiong Jie Li
- Department of Plastic Surgery, People's Hospital of Zhongshan City, Zhongshan, 528421, Guangdong, China
| | - Yongshi Ma
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Zhiming Mai
- Obstetrics and Gynaecology Department, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China
| | - Yao Yang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Min Luo
- Obstetrics and Gynaecology Department, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China
| | - Wenping Xu
- Department of General Practice, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China
| | - Kui Chen
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, Guangdong, China
- Obstetrics and Gynaecology Department, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China
| | - Xuri Chen
- Department of General Practice, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jianbing Tang
- Department of Burn and Plastic Surgery, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China
| | - Biao Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China.
- The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area of Chinese PLA, Guangzhou, 510010, China.
| | - Xiao Cui
- Department of Burn and Plastic Surgery, General Hospital of Southern Theatre Command, PLA, Guangzhou, 510010, China.
- Department of Physiotherapy, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
10
|
Chaudhary PK, Kim S, Kim S. Shedding Light on the Cell Biology of Platelet-Derived Extracellular Vesicles and Their Biomedical Applications. Life (Basel) 2023; 13:1403. [PMID: 37374185 DOI: 10.3390/life13061403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
EVs are membranous subcellular structures originating from various cells, including platelets which consist of biomolecules that can modify the target cell's pathophysiological functions including inflammation, cell communication, coagulation, and metastasis. EVs, which are known to allow the transmission of a wide range of molecules between cells, are gaining popularity in the fields of subcellular treatment, regenerative medicine, and drug delivery. PEVs are the most abundant EVs in circulation, being produced by platelet activation, and are considered to have a significant role in coagulation. PEV cargo is extremely diverse, containing lipids, proteins, nucleic acids, and organelles depending on the condition that induced their release and can regulate a wide range of biological activities. PEVs, unlike platelets, can overcome tissue barriers, allowing platelet-derived contents to be transferred to target cells and organs that platelets cannot reach. Their isolation, characterization, and therapeutic efficacy, on the other hand, are poorly understood. This review summarizes the technical elements of PEV isolation and characterization methods as well as the pathophysiological role of PEVs, including therapeutic potential and translational possibility in diverse disciplines.
Collapse
Affiliation(s)
- Preeti Kumari Chaudhary
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sanggu Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
11
|
Mesenchymal stem cells support human vascular endothelial cells to form vascular sprouts in human platelet lysate-based matrices. PLoS One 2022; 17:e0278895. [PMID: 36520838 PMCID: PMC9754269 DOI: 10.1371/journal.pone.0278895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
During tissue regeneration, mesenchymal stem cells can support endothelial cells in the process of new vessel formation. For a functional interaction of endothelial cells with mesenchymal stem cells a vascular inductive microenvironment is required. Using a cellular model for neo-vessel formation, we could show that newly formed vascular structures emanated from the embedded aggregates, consisting of mesenchymal stem cells co-cultured with autologous human umbilical vein endothelial cells, into avascular human platelet lysate-based matrices, bridging distances up to 5 mm to join with adjacent aggregates with the same morphology forming an interconnected network. These newly formed vascular sprouts showed branch points and generated a lumen, as sign of mature vascular development. In two-dimensional culture, we detected binding of mesenchymal stem cells to laser-damaged endothelial cells under flow conditions, mimicking the dynamics in blood vessels. In conclusion, we observed that mesenchymal stem cells can support human umbilical vein endothelial cells in their vitality and functionality. In xeno-free human platelet lysate-based matrices, endothelial cells form complex vascular networks in a primarily avascular scaffold with the aid of mesenchymal stem cells, when co-cultured in three-dimensional spherical aggregates. Under dynamic conditions, representing the flow rate of venous vessel, mesenchymal stem cells preferably bind to damaged endothelial cells presumably assisting in the healing process.
Collapse
|
12
|
Everts PA, Mazzola T, Mautner K, Randelli PS, Podesta L. Modifying Orthobiological PRP Therapies Are Imperative for the Advancement of Treatment Outcomes in Musculoskeletal Pathologies. Biomedicines 2022; 10:biomedicines10112933. [PMID: 36428501 PMCID: PMC9687216 DOI: 10.3390/biomedicines10112933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Autologous biological cellular preparations have materialized as a growing area of medical advancement in interventional (orthopedic) practices and surgical interventions to provide an optimal tissue healing environment, particularly in tissues where standard healing is disrupted and repair and ultimately restoration of function is at risk. These cellular therapies are often referred to as orthobiologics and are derived from patient's own tissues to prepare point of care platelet-rich plasma (PRP), bone marrow concentrate (BMC), and adipose tissue concentrate (ATC). Orthobiological preparations are biological materials comprised of a wide variety of cell populations, cytokines, growth factors, molecules, and signaling cells. They can modulate and influence many other resident cells after they have been administered in specific diseased microenvironments. Jointly, the various orthobiological cell preparations are proficient to counteract persistent inflammation, respond to catabolic reactions, and reinstate tissue homeostasis. Ultimately, precisely delivered orthobiologics with a proper dose and bioformulation will contribute to tissue repair. Progress has been made in understanding orthobiological technologies where the safety and relatively easy manipulation of orthobiological treatment tools has been demonstrated in clinical applications. Although more positive than negative patient outcome results have been registered in the literature, definitive and accepted standards to prepare specific cellular orthobiologics are still lacking. To promote significant and consistent clinical outcomes, we will present a review of methods for implementing dosing strategies, using bioformulations tailored to the pathoanatomic process of the tissue, and adopting variable preparation and injection volume policies. By optimizing the dose and specificity of orthobiologics, local cellular synergistic behavior will increase, potentially leading to better pain killing effects, effective immunomodulation, control of inflammation, and (neo) angiogenesis, ultimately contributing to functionally restored body movement patterns.
Collapse
Affiliation(s)
- Peter A. Everts
- Education & Research Division, Gulf Coast Biologics, Fort Myers, FL 33916, USA
- Correspondence: ; Tel.: +1-239-961-6457
| | - Timothy Mazzola
- Breakthrough Regenerative Orthopedics, Boulder, CO 80305, USA
| | - Kenneth Mautner
- Department of Physical Medicine and Rehabilitation, Emory University, Atlanta, GA 30329, USA
| | - Pietro S. Randelli
- Instituto Orthopedico Gaetano Pini, Milan University, 20122 Milan, Italy
| | | |
Collapse
|
13
|
Buzalaf MAR, Levy FM. Autologous platelet concentrates for facial rejuvenation. J Appl Oral Sci 2022; 30:e20220020. [PMID: 36074433 PMCID: PMC9444188 DOI: 10.1590/1678-7757-2022-0020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/02/2022] [Indexed: 12/04/2022] Open
Abstract
Autologous platelet concentrates (APCs) are promising therapeutic agents in facial rejuvenation since they are a great source of cytokines, growth factors and other biologically active substances. Obtained from the patient’s blood, they have the advantages of reducing immunological reactions, making the procedure safer, well tolerated, with minimal adverse effects and lower cost. Currently, they are used for facial rejuvenation both in combination with microneedling and in mesotherapy techniques, as well as to treat facial acne scars, melasma and wounds after laser ablative treatments. This review summarizes current knowledge on the use of APCs, ranging from basic concepts related to their composition and mechanisms of action to up-to-date information on their clinical efficacy.
Collapse
Affiliation(s)
- Marília Afonso Rabelo Buzalaf
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Flávia Mauad Levy
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| |
Collapse
|
14
|
Shi X, Li H, Xu Y, Nyalali AMK, Li F. The prognostic value of the preoperative inflammatory index on the survival of glioblastoma patients. Neurol Sci 2022; 43:5523-5531. [PMID: 35606674 PMCID: PMC9126244 DOI: 10.1007/s10072-022-06158-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The growth and development of tumors are closely related to the initiation and amplification of the inflammatory response. Various inflammatory biomarkers had attained growing attention for nearly two decades and were discovered strongly associated with cancer patients' prognosis, indicating that systemic inflammatory response is possibly essential to cancer progression. However, little was known about the sensitive biomarkers associated with the detection, persistence, treatment, and prognosis of GBM. Hence, the retrospective research endeavored to evaluate the prognostic value of preoperative inflammatory biomarkers in patients with GBM who initially received standardized treatment. METHODS The 232 glioblastoma patients eligible who were admitted to Qilu Hospitals in Shandong Province from January 2014 to January 2018 were collected for this analysis. Inflammatory markers, including the systemic immune-inflammation index (SII), systemic immune response index (SIRI), neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), monocyte-lymphocyte ratio (MLR), and albumin/globulin ratio (AGR), were designed. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method, and we calculated the area under the ROC curve to determine the AUC value. Besides, we used the Cox proportional hazard model to estimate the relationship between variables and PFS and OS. The statistical differences between variables and PFS and OS were tested through the log-rank test. What is more, the LR method was used to perform Cox multiple regression analysis. The results were represented by hazard ratio (HR), 95% CI, any 2-tailed P < 0.01 was accepted as statistically different. RESULTS The multivariate Cox proportional hazard model presented that SII ≥ 659.1 was an independent risk factor affecting OS (HR = 2.238, 95% CI = 1.471-3.406, P < 0.001) and postoperative PFS (HR = 2.000, 95% CI = 1.472-2.716, P < 0.001) in GBM patients. The 1-, 3-, and 5-year OS of the SII < 659.1 group was 70.8%, 26.9%, and 14.1%, respectively, while the 1- and 3-year OS of the SII ≥ 659.1 group was 37.5% and 11.5% (P < 0.001). The 1-, 3-, and 5-year PFS of the SII < 659.1 group was 36.3%, 19.6%, and 13%, respectively, while the 1-year PFS of the SII ≥ 659.1 group was 11.3% (P < 0.001). Results of patients' clinical and pathological characteristics paraded that in comparison to the lower SII group, the higher SII group had significantly inferior Karnofsky Performance Scale (KPS) scores (P < 0.001) and more frequent cystic changes of the tumors (P < 0.001), whereas the values of SIRI, NLR, PLR, MLR, and AGR were low. CONCLUSIONS SII is an independent inflammatory indicator for predicting the prognosis of GBM patients after receiving initially standardized treatments.
Collapse
Affiliation(s)
- Xiaohan Shi
- School of Nursing and Rehabilitation, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong Province, China
| | - Huayu Li
- School of Nursing and Rehabilitation, Shandong University, No. 44 Wenhua Xi Road, Jinan, 250012, Shandong Province, China
| | - Yongxiang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road, Jinan, 250012, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Alphonce M K Nyalali
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road, Jinan, 250012, China.,Department of Surgery, Songwe Regional Referral Hospital, Songwe Box 23, Mbeya, Tanzania.,Department of Orthopedics and Neurosurgery, Mbeya Zonal Referral Hospital and Mbeya College of Health and Allied Sciences, University of Dar Es Salaam, PO Box 419, Mbeya, Tanzania
| | - Feng Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, Wenhua Xi Road, Jinan, 250012, China. .,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China. .,Department of Neurosurgery, Shandong First Medical University Affiliated Cancer Hospital, Jiyan Road, Jinan, 250117, Shandong Province, China. .,Qilu Medical College of Shandong University, Wenhua Xi Road, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
15
|
Cytokine pathway variants modulate platelet production: IFNA16 is a thrombocytosis susceptibility locus in humans. Blood Adv 2022; 6:4884-4900. [PMID: 35381074 PMCID: PMC9631663 DOI: 10.1182/bloodadvances.2021005648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory stimuli have divergent effects on peripheral platelet counts, although the mechanisms of thrombocytopenic and thrombocytotic responses remain poorly understood. A candidate gene approach targeting 326 polymorphic genes enriched in thrombopoietic and cytokine signaling pathways was applied to identify single nucleotide variants (SNVs) implicated in enhanced platelet responses in cohorts with reactive thrombocytosis (RT) or essential (myeloproliferative neoplasm [MPN]) thrombocytosis (ET). Cytokine profiles incorporating a 15-member subset, pathway topology, and functional interactive networks were distinct between ET and RT, consistent with distinct regulatory pathways of exaggerated thrombopoiesis. Genetic studies using aggregate (ET + RT) or ET-restricted cohorts identified associations with 2 IFNA16 (interferon-α16) SNVs, and the ET associations were validated in a second independent cohort (P = .0002). Odds ratio of the combined ET cohort (n = 105) was 4.92, restricted to the JAK2V617F-negative subset (odds ratio, 5.01). ET substratification analysis by variant IFNA16 exhibited a statistically significant increase in IFN-α16 levels (P = .002) among 16 quantifiable cytokines. Recombinantly expressed variant IFN-α16 encompassing 3 linked non-synonymous SNVs (E65H95P133) retained comparable antiviral and pSTAT signaling profiles as native IFN-α16 (V65D95A133) or IFN-α2, although both native and variant IFN-α16 showed stage-restricted differences (compared with IFN-α2) of IFN-regulated genes in CD34+-stimulated megakaryocytes. These data implicate IFNA16 (IFN-α16 gene product) as a putative susceptibility locus (driver) within the broader disrupted cytokine network evident in MPNs, and they provide a framework for dissecting functional interactive networks regulating stress or MPN thrombopoiesis.
Collapse
|
16
|
The Growth Factor Release from a Platelet-Rich Plasma Preparation Is Influenced by the Onset of Guttate Psoriasis: A Case Report. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12147250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The involvement of platelets in immune and inflammatory processes is generally recognized; nevertheless, in psoriasis, their role is not clearly understood. We studied the in vitro growth factor release from a platelet-rich plasma preparation, the concentrated growth factors (CGF), in a case of a psoriasis subject three days before the onset of the papule. The CGF clots were incubated in a cell culture medium without growth supplements for 5 h and 1, 3, 6, 7, and 8 days, and the release kinetics of PDGF-AB, VEGF, TNF-α, and TGF-β1 were evaluated. The data, based on the results obtained during the case study, report a general increase in growth factor release in the psoriasis subject with respect to the healthy control, indicating an imbalance of growth factor production from blood cells. Although the results should be validated in the future, they show new aspects of this dermatological pathology, opening new possibilities both as the method of study, using CGF, and the involvement of platelets and growth factors in its development and maintenance.
Collapse
|
17
|
Nebie O, Buée L, Blum D, Burnouf T. Can the administration of platelet lysates to the brain help treat neurological disorders? Cell Mol Life Sci 2022; 79:379. [PMID: 35750991 PMCID: PMC9243829 DOI: 10.1007/s00018-022-04397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Neurodegenerative disorders of the central nervous system (CNS) and brain traumatic insults are characterized by complex overlapping pathophysiological alterations encompassing neuroinflammation, alterations of synaptic functions, oxidative stress, and progressive neurodegeneration that eventually lead to irreversible motor and cognitive dysfunctions. A single pharmacological approach is unlikely to provide a complementary set of molecular therapeutic actions suitable to resolve these complex pathologies. Recent preclinical data are providing evidence-based scientific rationales to support biotherapies based on administering neurotrophic factors and extracellular vesicles present in the lysates of human platelets collected from healthy donors to the brain. Here, we present the most recent findings on the composition of the platelet proteome that can activate complementary signaling pathways in vivo to trigger neuroprotection, synapse protection, anti-inflammation, antioxidation, and neurorestoration. We also report experimental data where the administration of human platelet lysates (HPL) was safe and resulted in beneficial neuroprotective effects in established rodent models of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, traumatic brain injury, and stroke. Platelet-based biotherapies, prepared from collected platelet concentrates (PC), are emerging as a novel pragmatic and accessible translational therapeutic strategy for treating neurological diseases. Based on this assumption, we further elaborated on various clinical, manufacturing, and regulatory issues that need to be addressed to ensure the ethical supply, quality, and safety of HPL preparations for treating neurodegenerative and traumatic pathologies of the CNS. HPL made from PC may become a unique approach for scientifically based treatments of neurological disorders readily accessible in low-, middle-, and high-income countries.
Collapse
Affiliation(s)
- Ouada Nebie
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France.
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France.
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thierry Burnouf
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre, Taipei Medical University Shuang-Ho Hospital, New Taipei City, 23561, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Wu S, Guo W, Li R, Zhang X, Qu W. Progress of Platelet Derivatives for Cartilage Tissue Engineering. Front Bioeng Biotechnol 2022; 10:907356. [PMID: 35782516 PMCID: PMC9243565 DOI: 10.3389/fbioe.2022.907356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Articular cartilage has limited self-regeneration ability for lacking of blood vessels, nerves, and lymph that makes it a great challenge to repair defects of the tissue and restore motor functions of the injured or aging population. Platelet derivatives, such as platelet-rich plasma, have been proved effective, safe, and economical in musculoskeletal diseases for their autologous origin and rich in growth factors. The combination of platelet derivatives with biomaterials provides both mechanical support and localized sustained release of bioactive molecules in cartilage tissue engineering and low-cost efficient approaches of potential treatment. In this review, we first provide an overview of platelet derivatives and their application in clinical and experimental therapies, and then we further discuss the techniques of the addition of platelet derivatives and their influences on scaffold properties. Advances in cartilage tissue engineering with platelet derivatives as signal factors and structural components are also introduced before prospects and concerns in this research field. In short, platelet derivatives have broad application prospects as an economical and effective enhancement for tissue engineering–based articular cartilage repair.
Collapse
Affiliation(s)
- Siyu Wu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Wenlai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xi Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| | - Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xi Zhang, ; Wenrui Qu,
| |
Collapse
|
19
|
Szelenberger R, Jóźwiak P, Kacprzak M, Bijak M, Zielińska M, Olender A, Saluk-Bijak J. Variations in Blood Platelet Proteome and Transcriptome Revealed Altered Expression of Transgelin-2 in Acute Coronary Syndrome Patients. Int J Mol Sci 2022; 23:ijms23116340. [PMID: 35683019 PMCID: PMC9181388 DOI: 10.3390/ijms23116340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 02/05/2023] Open
Abstract
Proteomic analyses based on mass spectrometry provide a powerful tool for the simultaneous identification of proteins and their signatures. Disorders detection at the molecular level delivers an immense impact for a better understanding of the pathogenesis and etiology of various diseases. Acute coronary syndrome (ACS) refers to a group of heart diseases generally associated with rupture of an atherosclerotic plaque and partial or complete thrombotic obstruction of the blood flow in the infarct-related coronary artery. The essential role in the pathogenesis of ACS is related to the abnormal, pathological activation of blood platelets. The multifactorial and complex character of ACS indicates the need to explain the molecular mechanisms responsible for thrombosis. In our study, we performed screening and comparative analysis of platelet proteome from ACS patients and healthy donors. Two-dimensional fluorescence difference gel electrophoresis and nanoscale liquid chromatography coupled to tandem mass spectrometry showed altered expressions of six proteins (i.e., vinculin, transgelin-2, fibrinogen β and γ chains, apolipoprotein a1, and tubulin β), with the overlapping increased expression at the mRNA level for transgelin-2. Dysregulation in protein expression identified in our study may be associated with an increased risk of thrombotic events, correlated with a higher aggregability of blood platelets and induced shape change, thus explaining the phenomenon of the hyperreactivity of blood platelets in ACS.
Collapse
Affiliation(s)
- Rafał Szelenberger
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
- Correspondence:
| | - Paweł Jóźwiak
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Michał Kacprzak
- Department of Interventional Cardiology, Medical University of Lodz, 91-213 Lodz, Poland; (M.K.); (M.Z.)
| | - Michał Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Marzenna Zielińska
- Department of Interventional Cardiology, Medical University of Lodz, 91-213 Lodz, Poland; (M.K.); (M.Z.)
| | - Alina Olender
- Chair and Department of Medical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
20
|
Chaudhary PK, Kim S, Kim S. An Insight into Recent Advances on Platelet Function in Health and Disease. Int J Mol Sci 2022; 23:ijms23116022. [PMID: 35682700 PMCID: PMC9181192 DOI: 10.3390/ijms23116022] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Platelets play a variety of roles in vascular biology and are best recognized as primary hemostasis and thrombosis mediators. Platelets have a large number of receptors and secretory molecules that are required for platelet functionality. Upon activation, platelets release multiple substances that have the ability to influence both physiological and pathophysiological processes including inflammation, tissue regeneration and repair, cancer progression, and spreading. The involvement of platelets in the progression and seriousness of a variety of disorders other than thrombosis is still being discovered, especially in the areas of inflammation and the immunological response. This review represents an integrated summary of recent advances on the function of platelets in pathophysiology that connects hemostasis, inflammation, and immunological response in health and disease and suggests that antiplatelet treatment might be used for more than only thrombosis.
Collapse
|
21
|
Chen X, Xu Y, Chen Q, Zhang H, Zeng Y, Geng Y, Shen L, Li F, Chen L, Chen GQ, Huang C, Liu J. The phosphatase PTEN links platelets with immune regulatory functions of mouse T follicular helper cells. Nat Commun 2022; 13:2762. [PMID: 35589797 PMCID: PMC9120038 DOI: 10.1038/s41467-022-30444-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/30/2022] [Indexed: 01/10/2023] Open
Abstract
Beyond a function in hemostasis and thrombosis, platelets can regulate innate and adaptive immune responses. Hyperactive platelets are frequently associated with multiple human autoimmune diseases, yet their pathogenic functions in these diseases have not been fully established. Emerging studies show an essential function of the phosphatase and tensin homolog (PTEN) in maintenance of immune homeostasis. Here, we show that mice with platelet-specific deletion of Pten, develop age-related lymphoproliferative diseases and humoral autoimmunity not seen in wildtype animals. Platelet-specific Pten-deficient mice have aberrant T cell activation, excessive T follicular helper (Tfh) cell responses and accumulation of platelet aggregates in lymph nodes. Transferred Pten-deficient platelets are able to infiltrate into the peripheral lymphoid tissues and form more aggregates. Moreover, Pten-deficient platelets are hyperactive and overproduce multiple Tfh-promoting cytokines via activation of the PDK1/mTORC2-AKT-SNAP23 pathway. Pten-deficient platelets show enhanced interaction with CD4+ T cells and promote conversion of CD4+ T cells into Tfh cells. Our results implicate PTEN in platelet-mediated immune homeostasis, and provide evidence that hyperactive platelets function as an important mediator in autoimmune diseases using mouse models.
Collapse
Affiliation(s)
- Xue Chen
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, China.
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qidi Chen
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Heng Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yu Zeng
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yan Geng
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Lei Shen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Fubin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Lei Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Guo-Qiang Chen
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Chuanxin Huang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
22
|
Uncomplicated Plasmodium vivax malaria: mapping the proteome from circulating platelets. Clin Proteomics 2022; 19:1. [PMID: 34991449 PMCID: PMC8903537 DOI: 10.1186/s12014-021-09337-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 12/21/2021] [Indexed: 01/07/2023] Open
Abstract
Background Thrombocytopenia is frequent in Plasmodium vivax malaria but the role of platelets in pathogenesis is unknown. Our study explores the platelet (PLT) proteome from uncomplicated P. vivax patients, to fingerprint molecular pathways related to platelet function. Plasma levels of Platelet factor 4 (PF4/CXCL4) and Von Willebrand factor (VWf), as well as in vitro PLTs—P. vivax infected erythrocytes (Pv-IEs) interactions were also evaluated to explore the PLT response and effect on parasite development. Methods A cohort of 48 patients and 25 healthy controls were enrolled. PLTs were purified from 5 patients and 5 healthy controls for Liquid Chromatography–Mass spectrometry (LC–MS/MS) analysis. Plasma levels of PF4/CXCL4 and VWf were measured in all participants. Additionally, P. vivax isolates (n = 10) were co-cultured with PLTs to measure PLT activation by PF4/CXCL4 and Pv-IE schizonts formation by light microscopy. Results The proteome from uncomplicated P. vivax patients showed 26 out of 215 proteins significantly decreased. PF4/CXCL4 was significantly decreased followed by other proteins involved in platelet activation, cytoskeletal remodeling, and endothelial adhesion, including glycoprotein V that was significantly decreased in thrombocytopenic patients. In contrast, acute phase proteins, including SERPINs and Amyloid Serum A1 were increased. High levels of VWf in plasma from patients suggested endothelial activation while PF4/CXCL4 plasma levels were similar between patients and controls. Interestingly, high levels of PF4/CXCL4 were released from PLTs—Pv-IEs co-cultures while Pv-IEs schizont formation was inhibited. Conclusions The PLT proteome analyzed in this study suggests that PLTs actively respond to P. vivax infection. Altogether, our findings suggest important roles of PF4/CXCL4 during uncomplicated P. vivax infection through a possible intracellular localization. Our study shows that platelets are active responders to P. vivax infection, inhibiting intraerythrocytic parasite development. Future studies are needed to further investigate the molecular pathways of interaction between platelet proteins found in this study and host response, which could affect parasite control as well as disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09337-7.
Collapse
|
23
|
Rehman F, Krishan K, Latif I, Sudan E, Sultan J, Hassan I. Intra-individual Right-Left Comparative Study of Combined Therapy of Intramatricial Triamcinolone and Platelet-Rich Plasma vs. Intramatricial Triamcinolone Only in Lichen Planus-Associated Nail Dystrophy. J Cutan Aesthet Surg 2021; 14:311-317. [PMID: 34908773 PMCID: PMC8611714 DOI: 10.4103/jcas.jcas_156_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background While intramatricial triamcinolone is the most commonly used treatment option for onychodystrophy due to lichen planus, the role of platelet-rich plasma (PRP) in the treatment of onychodystrophy is not established. So we combined the two treatment modalities to assess the synergistic action on nail dystrophy. Aims and Objectives The aim of this article is to compare the effect of combination of intramatricial triamcinolone and PRP vs. intramatricial triamcinolone alone in the treatment of onychodystrophy. Materials and Methods This study was done in 26 and 25 pairs of symmetrical dystrophic nails in hands and feet, respectively. The patients received intramatricial injection of triamcinolone and PRP on one side of finger and toe nails and triamcinolone on the contralateral side. Results Statistically significant improvements in grades of dystrophy according to the Nail Dystrophy Grading System (NDGS) in the finger and toe nails treated with combined therapy were obtained (P < 0.001 and = 0.002, respectively). According to the physician global assessment, response to combination therapy was statistically significant between the two comparison groups of finger and toe nails (P = 0.001 and = 0.004, respectively). Similarly, according to the patient satisfaction score, statistically significant difference was found between the comparison groups of combination therapy and single therapy (P < 0.001). Conclusion Addition of PRP significantly improved the nail quality. Intramatricial PRP is a safe and effective therapeutic modality in refractory nail dystrophies.
Collapse
Affiliation(s)
- Fozia Rehman
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| | - Kewal Krishan
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| | - Insha Latif
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| | - Ekta Sudan
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| | - Javeed Sultan
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| | - Iffat Hassan
- Department of Dermatology, Venereology, and Leprosy, Government Medical College Srinagar, Karan Nagar, Jammu and Kashmir, India
| |
Collapse
|
24
|
Alexander S, Horo I, Johnson S, Daniel S. Platelet-rich plasma in hair loss-Mechanism, preparation, and classification. J Cosmet Dermatol 2021; 21:970-978. [PMID: 34897939 DOI: 10.1111/jocd.14662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Abstract
Platelet-rich plasma is the autologous plasma containing platelet concentration more than the baseline separated after centrifugation. It has become a very popular option for the treatment of hair loss in the past few decades. Used alone or in combination with other treatment options it has found a place as a hair restoration procedure throughout the world. However, evidence supporting its credibility is quite ambiguous with contradictory reports available in the literature. In this article, we try to analyze the available data regarding its mechanism of action, preparation protocols, and classification, with regard to hair loss.
Collapse
Affiliation(s)
- Sajin Alexander
- Schieffelin Institute of Health-Research and Leprosy Center (SIH-R &LC), Karigiri, Vellore, India
| | - Ilse Horo
- Schieffelin Institute of Health-Research and Leprosy Center (SIH-R &LC), Karigiri, Vellore, India
| | - Sobha Johnson
- Schieffelin Institute of Health-Research and Leprosy Center (SIH-R &LC), Karigiri, Vellore, India
| | - Sheela Daniel
- Schieffelin Institute of Health-Research and Leprosy Center (SIH-R &LC), Karigiri, Vellore, India
| |
Collapse
|
25
|
Cognasse F, Hamzeh-Cognasse H, Mismetti P, Thomas T, Eglin D, Marotte H. The Non-Haemostatic Response of Platelets to Stress: An Actor of the Inflammatory Environment on Regenerative Medicine? Front Immunol 2021; 12:741988. [PMID: 34589090 PMCID: PMC8473800 DOI: 10.3389/fimmu.2021.741988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | | | - Patrick Mismetti
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Etienne, France
| | - Thierry Thomas
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Department of Rheumatology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - David Eglin
- Mines Saint-Étienne, Univ Lyon, Univ Jean Monnet, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | - Hubert Marotte
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Department of Rheumatology, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
26
|
Jansen EE, Braun A, Jansen P, Hartmann M. Platelet-Therapeutics to Improve Tissue Regeneration and Wound Healing-Physiological Background and Methods of Preparation. Biomedicines 2021; 9:biomedicines9080869. [PMID: 34440073 PMCID: PMC8389548 DOI: 10.3390/biomedicines9080869] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/18/2022] Open
Abstract
Besides their function in primary hemostasis, platelets are critically involved in the physiological steps leading to wound healing and tissue repair. For this purpose, platelets have a complex set of receptors allowing the recognition, binding, and manipulation of extracellular structures and the detection of pathogens and tissue damage. Intracellular vesicles contain a huge set of mediators that can be released to the extracellular space to coordinate the action of platelets as other cell types for tissue repair. Therapeutically, the most frequent use of platelets is the intravenous application of platelet concentrates in case of thrombocytopenia or thrombocytopathy. However, there is increasing evidence that the local application of platelet-rich concentrates and platelet-rich fibrin can improve wound healing and tissue repair in various settings in medicine and dentistry. For the therapeutic use of platelets in wound healing, several preparations are available in clinical practice. In the present study we discuss the physiology and the cellular mechanisms of platelets in hemostasis and wound repair, the methods used for the preparation of platelet-rich concentrates and platelet-rich fibrin, and highlight some examples of the therapeutic use in medicine and dentistry.
Collapse
Affiliation(s)
- Ellen E. Jansen
- Clinic for Operative Dentistry, Periodontology and Preventive Dentistry, RWTH Aachen University, 52074 Aachen, Germany; (E.E.J.); (A.B.); (P.J.)
| | - Andreas Braun
- Clinic for Operative Dentistry, Periodontology and Preventive Dentistry, RWTH Aachen University, 52074 Aachen, Germany; (E.E.J.); (A.B.); (P.J.)
| | - Patrick Jansen
- Clinic for Operative Dentistry, Periodontology and Preventive Dentistry, RWTH Aachen University, 52074 Aachen, Germany; (E.E.J.); (A.B.); (P.J.)
| | - Matthias Hartmann
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, 45122 Essen, Germany
- Correspondence:
| |
Collapse
|
27
|
Chaudhary PK, Kim S. The GRKs Reactome: Role in Cell Biology and Pathology. Int J Mol Sci 2021; 22:ijms22073375. [PMID: 33806057 PMCID: PMC8036551 DOI: 10.3390/ijms22073375] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor kinases (GRKs) are protein kinases that function in concert with arrestins in the regulation of a diverse class of G protein-coupled receptors (GPCRs) signaling. Although GRKs and arrestins are key participants in the regulation of GPCR cascades, the complex regulatory mechanisms of GRK expression, its alternation, and their function are not thoroughly understood. Several studies together with the work from our lab in recent years have revealed the critical role of these kinases in various physiological and pathophysiological processes, including cardiovascular biology, inflammation and immunity, neurodegeneration, thrombosis, and hemostasis. A comprehensive understanding of the mechanisms underlying functional interactions with multiple receptor proteins and how these interactions take part in the development of various pathobiological processes may give rise to novel diagnostic and therapeutic strategies. In this review, we summarize the current research linking the role of GRKs to various aspects of cell biology, pathology, and therapeutics, with a particular focus on thrombosis and hemostasis.
Collapse
|
28
|
Seo SS, Lee IS, Lee GH. Intra-articular Injection Therapy and Biologic Treatment. A STRATEGIC APPROACH TO KNEE ARTHRITIS TREATMENT 2021:171-212. [DOI: 10.1007/978-981-16-4217-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
29
|
Balkenhol J, Kaltdorf KV, Mammadova-Bach E, Braun A, Nieswandt B, Dittrich M, Dandekar T. Comparison of the central human and mouse platelet signaling cascade by systems biological analysis. BMC Genomics 2020; 21:897. [PMID: 33353544 PMCID: PMC7756956 DOI: 10.1186/s12864-020-07215-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background Understanding the molecular mechanisms of platelet activation and aggregation is of high interest for basic and clinical hemostasis and thrombosis research. The central platelet protein interaction network is involved in major responses to exogenous factors. This is defined by systemsbiological pathway analysis as the central regulating signaling cascade of platelets (CC). Results The CC is systematically compared here between mouse and human and major differences were found. Genetic differences were analysed comparing orthologous human and mouse genes. We next analyzed different expression levels of mRNAs. Considering 4 mouse and 7 human high-quality proteome data sets, we identified then those major mRNA expression differences (81%) which were supported by proteome data. CC is conserved regarding genetic completeness, but we observed major differences in mRNA and protein levels between both species. Looking at central interactors, human PLCB2, MMP9, BDNF, ITPR3 and SLC25A6 (always Entrez notation) show absence in all murine datasets. CC interactors GNG12, PRKCE and ADCY9 occur only in mice. Looking at the common proteins, TLN1, CALM3, PRKCB, APP, SOD2 and TIMP1 are higher abundant in human, whereas RASGRP2, ITGB2, MYL9, EIF4EBP1, ADAM17, ARRB2, CD9 and ZYX are higher abundant in mouse. Pivotal kinase SRC shows different regulation on mRNA and protein level as well as ADP receptor P2RY12. Conclusions Our results highlight species-specific differences in platelet signaling and points of specific fine-tuning in human platelets as well as murine-specific signaling differences. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-020-07215-4.
Collapse
Affiliation(s)
- Johannes Balkenhol
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany
| | - Kristin V Kaltdorf
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany
| | - Elmina Mammadova-Bach
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Centre, University of Würzburg, Würzburg, Germany.,Present address: Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig, Maximilian University of Munich, D-80336, Munich, Germany
| | - Attila Braun
- Member of the German Center for Lung Research (DZL), Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Centre, University of Würzburg, Würzburg, Germany
| | - Marcus Dittrich
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany.,Dept of Genetics, Biocenter, Am Hubland, University of Würzburg, Am Hubland, D 97074, Würzburg, Germany
| | - Thomas Dandekar
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany.
| |
Collapse
|
30
|
Kyselova A, Siragusa M, Anthes J, Solari FA, Loroch S, Zahedi RP, Walter U, Fleming I, Randriamboavonjy V. Cyclin Y is expressed in Platelets and Modulates Integrin Outside-in Signaling. Int J Mol Sci 2020; 21:ijms21218239. [PMID: 33153214 PMCID: PMC7662234 DOI: 10.3390/ijms21218239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
Diabetes is associated with platelet hyper-reactivity and enhanced risk of thrombosis development. Here we compared protein expression in platelets from healthy donors and diabetic patients to identify differentially expressed proteins and their possible function in platelet activation. Mass spectrometry analyses identified cyclin Y (CCNY) in platelets and its reduced expression in platelets from diabetic patients, a phenomenon that could be attributed to the increased activity of calpains. To determine the role of CCNY in platelets, mice globally lacking the protein were studied. CCNY-/- mice demonstrated lower numbers of circulating platelets but platelet responsiveness to thrombin and a thromboxane A2 analogue were comparable with that of wild-type mice, as was agonist-induced α and dense granule secretion. CCNY-deficient platelets demonstrated enhanced adhesion to fibronectin and collagen as well as an attenuated spreading and clot retraction, indicating an alteration in "outside in" integrin signalling. This phenotype was accompanied by a significant reduction in the agonist-induced tyrosine phosphorylation of β3 integrin. Taken together we have shown that CCNY is present in anucleated platelets where it is involved in the regulation of integrin-mediated outside in signalling associated with thrombin stimulation.
Collapse
Affiliation(s)
- Anastasia Kyselova
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, 60590 Frankfurt, Germany; (A.K.); (M.S.); (J.A.); (I.F.)
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
| | - Mauro Siragusa
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, 60590 Frankfurt, Germany; (A.K.); (M.S.); (J.A.); (I.F.)
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
| | - Julian Anthes
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, 60590 Frankfurt, Germany; (A.K.); (M.S.); (J.A.); (I.F.)
| | - Fiorella Andrea Solari
- Leibniz–Institute for Analytical Sciences (ISAS)- e.V., Otto-Hahn-Str. 6b, 44227 Dortmund, Germany;
| | - Stefan Loroch
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
- Leibniz–Institute for Analytical Sciences (ISAS)- e.V., Otto-Hahn-Str. 6b, 44227 Dortmund, Germany;
| | - René P. Zahedi
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
- Leibniz–Institute for Analytical Sciences (ISAS)- e.V., Otto-Hahn-Str. 6b, 44227 Dortmund, Germany;
| | - Ulrich Walter
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, 55131 Mainz, Germany
| | - Ingrid Fleming
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, 60590 Frankfurt, Germany; (A.K.); (M.S.); (J.A.); (I.F.)
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signaling, Centre of Molecular Medicine, Goethe University, Frankfurt am Main, 60590 Frankfurt, Germany; (A.K.); (M.S.); (J.A.); (I.F.)
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, 17475 Greifswald, Germany; (S.L.); (R.P.Z.); (U.W.)
- Correspondence: ; Tel.: +49-69-6301-6973; Fax: +49-69-6301-86880
| |
Collapse
|
31
|
Sims MC, Mayer L, Collins JH, Bariana TK, Megy K, Lavenu-Bombled C, Seyres D, Kollipara L, Burden FS, Greene D, Lee D, Rodriguez-Romera A, Alessi MC, Astle WJ, Bahou WF, Bury L, Chalmers E, Da Silva R, De Candia E, Deevi SVV, Farrow S, Gomez K, Grassi L, Greinacher A, Gresele P, Hart D, Hurtaud MF, Kelly AM, Kerr R, Le Quellec S, Leblanc T, Leinøe EB, Mapeta R, McKinney H, Michelson AD, Morais S, Nugent D, Papadia S, Park SJ, Pasi J, Podda GM, Poon MC, Reed R, Sekhar M, Shalev H, Sivapalaratnam S, Steinberg-Shemer O, Stephens JC, Tait RC, Turro E, Wu JKM, Zieger B, Kuijpers TW, Whetton AD, Sickmann A, Freson K, Downes K, Erber WN, Frontini M, Nurden P, Ouwehand WH, Favier R, Guerrero JA. Novel manifestations of immune dysregulation and granule defects in gray platelet syndrome. Blood 2020; 136:1956-1967. [PMID: 32693407 PMCID: PMC7582559 DOI: 10.1182/blood.2019004776] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Gray platelet syndrome (GPS) is a rare recessive disorder caused by biallelic variants in NBEAL2 and characterized by bleeding symptoms, the absence of platelet α-granules, splenomegaly, and bone marrow (BM) fibrosis. Due to the rarity of GPS, it has been difficult to fully understand the pathogenic processes that lead to these clinical sequelae. To discern the spectrum of pathologic features, we performed a detailed clinical genotypic and phenotypic study of 47 patients with GPS and identified 32 new etiologic variants in NBEAL2. The GPS patient cohort exhibited known phenotypes, including macrothrombocytopenia, BM fibrosis, megakaryocyte emperipolesis of neutrophils, splenomegaly, and elevated serum vitamin B12 levels. Novel clinical phenotypes were also observed, including reduced leukocyte counts and increased presence of autoimmune disease and positive autoantibodies. There were widespread differences in the transcriptome and proteome of GPS platelets, neutrophils, monocytes, and CD4 lymphocytes. Proteins less abundant in these cells were enriched for constituents of granules, supporting a role for Nbeal2 in the function of these organelles across a wide range of blood cells. Proteomic analysis of GPS plasma showed increased levels of proteins associated with inflammation and immune response. One-quarter of plasma proteins increased in GPS are known to be synthesized outside of hematopoietic cells, predominantly in the liver. In summary, our data show that, in addition to the well-described platelet defects in GPS, there are immune defects. The abnormal immune cells may be the drivers of systemic abnormalities such as autoimmune disease.
Collapse
Affiliation(s)
- Matthew C Sims
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Louisa Mayer
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Janine H Collins
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Tadbir K Bariana
- Department of Haematology, University of Cambridge, and
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Karyn Megy
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Cecile Lavenu-Bombled
- Assistance Publique-Hôpitaux de Paris, Centre de Reference des Pathologies Plaquettaires, Hôpitaux Armand Trousseau, Bicêtre, Robert Debré, Paris, France
| | - Denis Seyres
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | | | - Frances S Burden
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Daniel Greene
- Department of Haematology, University of Cambridge, and
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Forvie Site, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Dave Lee
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Antonio Rodriguez-Romera
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Marie-Christine Alessi
- Centre for CardioVascular and Nutrition Research, INSERM 1263, INRAE 1260, Marseille, France
| | - William J Astle
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Forvie Site, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Wadie F Bahou
- Department of Medicine, Stony Brook University, Stony Brook, NY
| | - Loredana Bury
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | | | - Rachael Da Silva
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Erica De Candia
- Institute of Internal Medicine and Geriatrics, Catholic University School of Medicine, Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Sri V V Deevi
- Department of Haematology, University of Cambridge, and
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Keith Gomez
- Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Andreas Greinacher
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Perugia, Italy
| | - Dan Hart
- The Royal London Hospital Haemophilia Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Marie-Françoise Hurtaud
- Assistance Publique-Hôpitaux de Paris, Centre de Reference des Pathologies Plaquettaires, Hôpitaux Armand Trousseau, Bicêtre, Robert Debré, Paris, France
| | - Anne M Kelly
- Department of Haematology, University of Cambridge, and
| | - Ron Kerr
- Department of Haematology, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Sandra Le Quellec
- Service d'Hématologie Biologique, Hospices Civils de Lyon, Lyon, France
| | - Thierry Leblanc
- Assistance Publique-Hôpitaux de Paris, Centre de Reference des Pathologies Plaquettaires, Hôpitaux Armand Trousseau, Bicêtre, Robert Debré, Paris, France
| | - Eva B Leinøe
- Department of Haematology, Rigshospitalet, Copenhagen, Denmark
| | - Rutendo Mapeta
- Department of Haematology, University of Cambridge, and
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Alan D Michelson
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Sara Morais
- Serviço de Hematologia Clínica, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Unidade Multidisciplinar de Investigação Biomédica, Instituto de Ciências Biomédicas, Universidade do Porto, Porto, Portugal
| | - Diane Nugent
- Center for Inherited Bleeding Disorders, Children's Hospital of Orange County, Orange, CA
| | - Sofia Papadia
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Soo J Park
- Division of Hematology and Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - John Pasi
- The Royal London Hospital Haemophilia Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gian Marco Podda
- Unità di Medicina 2, ASST Santi Paolo e Carlo, Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Man-Chiu Poon
- University of Calgary Cumming School of Medicine and Southern Alberta Rare Blood and Bleeding Disorders Comprehensive Care Program, Calgary, AB, Canada
| | - Rachel Reed
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Mallika Sekhar
- Department of Haematology, Royal Free London NHS Trust, London, United Kingdom
| | - Hanna Shalev
- Department of Pediatric Hematology/Oncology, Soroka Medical Center, Faculty of Medicine, Ben-Gurion University, Beer Sheva, Israel
| | - Suthesh Sivapalaratnam
- Department of Haematology, University of Cambridge, and
- Department of Haematology, Barts Health NHS Trust, London, United Kingdom
| | - Orna Steinberg-Shemer
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan C Stephens
- Department of Haematology, University of Cambridge, and
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Robert C Tait
- Department of Haematology, Royal Infirmary, Glasgow, United Kingdom
| | - Ernest Turro
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Forvie Site, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - John K M Wu
- Division of Hematology-Oncology, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Sanquin Research Institute, Department of Blood Cell Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Anthony D Whetton
- Stoller Biomarker Discovery Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e. V., Dortmund, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, United Kingdom
- Medizinische Fakultät, Medizinisches Proteom Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Kathleen Freson
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, University of Leuven, Leuven, Belgium
| | - Kate Downes
- Department of Haematology, University of Cambridge, and
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Wendy N Erber
- Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, Australia
- PathWest Laboratory Medicine, The University of Western Australia, Nedlands, Australia
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- British Heart Foundation, Cambridge Centre for Research Excellence, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Paquita Nurden
- Institut Hospitalo-Universitaire L'Institut de Rythmologie et Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; and
| | - Remi Favier
- Assistance Publique-Hôpitaux de Paris, Centre de Reference des Pathologies Plaquettaires, Hôpitaux Armand Trousseau, Bicêtre, Robert Debré, Paris, France
- INSERM Unité Mixte de Recherche 1170, Gustave Roussy Cancer Campus, Universite Paris-Saclay, Villejuif, France
| | - Jose A Guerrero
- Department of Haematology, University of Cambridge, and
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
32
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 waitfor delay '0:0:5'-- wvzy] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
33
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null-- rqgz] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
34
|
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
35
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null,null,null,null,null,null,null-- tbwa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
36
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 and sleep(5)-- larb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
37
|
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
38
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null,null-- wfik] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
39
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [PMID: 33096812 DOI: 10.3390/ijms21207794;select dbms_pipe.receive_message(chr(114)||chr(122)||chr(104)||chr(84),5) from dual--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
40
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 and 9425=(select 9425 from pg_sleep(5))-- untq] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
41
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 and sleep(5)] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
42
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 and 9425=(select 9425 from pg_sleep(5))] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
43
|
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
44
|
Everts P, Onishi K, Jayaram P, Lana JF, Mautner K. Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020; 21:ijms21207794. [PMID: 33096812 PMCID: PMC7589810 DOI: 10.3390/ijms21207794] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
Affiliation(s)
- Peter Everts
- Gulf Coast Biologics, Research and Science Division, Fort Myers, FL 33916, USA
- Correspondence: ; Tel.: +1-239-848-9555
| | - Kentaro Onishi
- Department of PM&R and Orthopedic Surgery, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA;
| | - Prathap Jayaram
- Department of Physical Medicine & Rehabilitation, Baylor College of Medicine, Houston, TX 77030, USA;
| | - José Fábio Lana
- The Bone and Cartilage Institute, Indaiatuba, Sao Paulo, Brazil;
| | - Kenneth Mautner
- Emory Sports Medicine and Primary Care Sports Medicine, Emory University, Atlanta, GA 30329, USA;
| |
Collapse
|
45
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null,null,null,null,null,null,null,null-- fsob] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
46
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null,null,null-- kwux] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
47
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null-- lozi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
48
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null-- krmy] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
49
|
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|
50
|
Platelet-Rich Plasma: New Performance Understandings and Therapeutic Considerations in 2020. Int J Mol Sci 2020. [DOI: 10.3390/ijms21207794 union all select null,null,null,null,null,null,null-- pkke] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Emerging autologous cellular therapies that utilize platelet-rich plasma (PRP) applications have the potential to play adjunctive roles in a variety of regenerative medicine treatment plans. There is a global unmet need for tissue repair strategies to treat musculoskeletal (MSK) and spinal disorders, osteoarthritis (OA), and patients with chronic complex and recalcitrant wounds. PRP therapy is based on the fact that platelet growth factors (PGFs) support the three phases of wound healing and repair cascade (inflammation, proliferation, remodeling). Many different PRP formulations have been evaluated, originating from human, in vitro, and animal studies. However, recommendations from in vitro and animal research often lead to different clinical outcomes because it is difficult to translate non-clinical study outcomes and methodology recommendations to human clinical treatment protocols. In recent years, progress has been made in understanding PRP technology and the concepts for bioformulation, and new research directives and new indications have been suggested. In this review, we will discuss recent developments regarding PRP preparation and composition regarding platelet dosing, leukocyte activities concerning innate and adaptive immunomodulation, serotonin (5-HT) effects, and pain killing. Furthermore, we discuss PRP mechanisms related to inflammation and angiogenesis in tissue repair and regenerative processes. Lastly, we will review the effect of certain drugs on PRP activity, and the combination of PRP and rehabilitation protocols.
Collapse
|