1
|
Bourrienne MC, Loyau S, Faille D, Gay J, Akhenak S, Farkh C, Ollivier V, Solonomenjanahary M, Dupont S, Choqueux C, Villeval JL, Plo I, Edmond V, Ho-Tin-Noé B, Ajzenberg N, Mazighi M. Impaired fibrinolysis in JAK2V617F-related myeloproliferative neoplasms. J Thromb Haemost 2024; 22:3199-3208. [PMID: 39260744 DOI: 10.1016/j.jtha.2024.07.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Myeloproliferative neoplasms (MPNs) are characterized by a high rate of thrombotic complications that contribute to morbidity and mortality. MPN-related thrombogenesis is assumed to be multifactorial, involving both procoagulant and proinflammatory processes. Whether impaired fibrinolysis also participates in the prothrombotic phenotype of MPN has been poorly investigated. OBJECTIVES We determined whether MPN, particularly JAK2V617F-positive MPN, is associated with fibrinolytic changes. METHODS Tissue-type plasminogen activator (tPA)-mediated fibrinolysis was evaluated both in whole blood and plasma from mice with a hematopoietic-restricted Jak2V617F expression compared with wild-type (WT) mice (Jak2WT) using (1) halo clot lysis, (2) front lysis, and (3) plasmin generation assays. tPA clot lysis assay was performed in the plasma from 65 MPN patients (JAK2V617F mutation, n = 50; CALR mutations, n = 9) compared with 28 healthy controls. RESULTS In whole blood from Jak2V617F mice, we observed a decreased fibrinolysis characterized by a significantly lower halo clot lysis rate compared with Jak2WT (95 ± 22 vs 147 ± 39 AU/min; P < .05). Similar results were observed in plasma (halo clot lysis rate, 130 ± 27 vs 186 ± 29 AU/min; front lysis rate, 2.8 ± 1.6 vs 6.1 ± 1.2 μm.min-1; P < .05). Plasmin generation was significantly decreased both in plasma clots and standardized fibrin clots from Jak2V617F mice compared with Jak2WT mice. Among MPN patients, impaired tPA-related fibrinolysis with prolonged clot lysis time was observed in JAK2V617F and CALR patients. Plasminogen activator inhibitor-1 and α2-antiplasmin were significantly increased in plasma from JAK2V617F patients compared with controls. CONCLUSION Our results suggest that impaired tPA-mediated fibrinolysis represents an important prothrombotic mechanism in MPN patients that requires confirmation in larger studies.
Collapse
Affiliation(s)
- Marie-Charlotte Bourrienne
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Laboratoire Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France.
| | - Stéphane Loyau
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
| | - Dorothée Faille
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Laboratoire Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Juliette Gay
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Laboratoire Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Séléna Akhenak
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
| | - Carine Farkh
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Laboratoire Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Véronique Ollivier
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France
| | | | - Sébastien Dupont
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France
| | - Christine Choqueux
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
| | - Jean-Luc Villeval
- INSERM U1287, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Valérie Edmond
- INSERM U1287, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Benoît Ho-Tin-Noé
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France
| | - Nadine Ajzenberg
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Laboratoire Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Mikaël Mazighi
- Optimisation Thérapeutique en Neuropsychopharmacologie, INSERM U1144, Université Paris Cité, Paris, France; Département de Neurologie, AP-HP, Hôpital Lariboisière, FHU NeuroVasc, Paris, France
| |
Collapse
|
2
|
Chen YY, Wang YH, Chen CC, Huang CE, Hsu CC, Hsiao SH, Leu YW. Exogenous Janus Kinase 617 Codon Influences Small Noncoding RNAs and Gene Expression in Ba/F3 Cells. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:344-354. [PMID: 39324984 DOI: 10.4103/ejpi.ejpi-d-24-00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/09/2024] [Indexed: 09/27/2024]
Abstract
ABSTRACT Myeloproliferative neoplasms (MPNs) are blood cancers caused by mutations that originate from hematopoietic stem cells. More than 50%-90% of MPN patients had a dominant negative valine (V) to phenylalanine (F) mutation at the Janus kinase 617 codon (JAK2V617F) within the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway; however, this mutation was also found in a high percentage of the general population, its penetrance varied, and its onset was shown to be polygenic. Consequently, it is still unknown what molecular mechanism underlies the MPN transformation produced by JAK2V617F. Patients with MPN have been shown to have dysregulation of noncoding RNAs, such as microRNA (miRNA) and PIWI-interacting RNA (piRNA), although there is not any concrete proof that JAK2V617F alone is responsible for the aberrant regulation of miRNA and piRNA. Human wild type versus V617F-mutated JAK2 are expressed in mouse Ba/F3 cells, and the expressed small and total RNAs were subjected to next generation sequencing analysis to determine the direct induction. Differentially expressed miRNAs, gene expression, and transcript and gene variations were found between exogenously expressed JAK2 and JAK2V617F in Ba/F3 cells. The differently expressed variations contained enriched transposable elements and piRNAs, indicating a rearranged epigenome. The results of the pathway analysis show that the transformation that self-validated the chosen sequencing target genes is impacted by the JAK-STAT pathway. The induction route is functionally conserved, according to exogenously produced miRNA and gene expression. These results may clarify how the JAK2V617F induces transformation.
Collapse
Affiliation(s)
- Yi-Yang Chen
- Division of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ying-Hsuan Wang
- Division of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Cheng Chen
- Division of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cih-En Huang
- Division of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chen Hsu
- Division of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Shu-Huei Hsiao
- Department of Biomedical Sciences, Institute of Molecular Biology and Institute of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Yu-Wei Leu
- Department of Biomedical Sciences, Institute of Molecular Biology and Institute of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
3
|
Reeves B. Blood cell JAKtivation aggravates cerebral venous thrombosis. Blood Adv 2024; 8:3327-3329. [PMID: 38916898 PMCID: PMC11258618 DOI: 10.1182/bloodadvances.2024012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024] Open
Affiliation(s)
- Brandi Reeves
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
4
|
Bourrienne MC, Le Cam Duchez V, Faille D, Farkh C, Solo Nomenjanahary M, Gay J, Loyau S, Journé C, Dupont S, Ollivier V, Villeval JL, Plo I, Edmond V, Jandrot-Perrus M, Labrouche-Colomer S, Cassinat B, Verger E, Desilles JP, Ho-Tin-Noé B, Triquenot Bagan A, Mazighi M, Ajzenberg N. Exacerbation of thromboinflammation by JAK2V617F mutation worsens the prognosis of cerebral venous sinus thrombosis. Blood Adv 2024; 8:3330-3343. [PMID: 38386979 PMCID: PMC11258627 DOI: 10.1182/bloodadvances.2023011692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
ABSTRACT Cerebral venous sinus thrombosis (CVST) is an uncommon venous thromboembolic event accounting for <1% of strokes resulting in brain parenchymal injuries. JAK2V617F mutation, the most frequent driving mutation of myeloproliferative neoplasms, has been reported to be associated with worse clinical outcomes in patients with CVST. We investigated whether hematopoietic JAK2V617F expression predisposes to specific pathophysiological processes and/or worse prognosis after CVST. Using an in vivo mouse model of CVST, we analyzed clinical, biological, and imaging outcomes in mice with hematopoietic-restricted Jak2V617F expression, compared with wild-type Jak2 mice. In parallel, we studied a human cohort of JAK2V617F-positive or -negative CVST. Early after CVST, mice with hematopoietic Jak2V617F expression had increased adhesion of platelets and neutrophils in cerebral veins located in the vicinity of CVST. On day 1, Jak2V617F mice had a worse outcome characterized by significantly more frequent and severe intracranial hemorrhages (ICHs) and higher mortality rates. Peripheral neutrophil activation was enhanced, as indicated by higher circulating platelet-neutrophil aggregates, upregulated CD11b expression, and higher myeloperoxydase plasma level. Concurrently, immunohistological and brain homogenate analysis showed higher neutrophil infiltration and increased blood-brain barrier disruption. Similarly, patients with JAK2V617F-positive CVST tended to present higher thrombotic burden and had significantly higher systemic immune-inflammation index, a systemic thromboinflammatory marker, than patients who were JAK2V617F-negative. In mice with CVST, our study corroborates that Jak2V617F mutation leads to a specific pattern including increased thrombotic burden, ICH, and mortality. The exacerbated thromboinflammatory response, observed both in mice and patients positive for JAK2V617F, could contribute to hemorrhagic complications.
Collapse
Affiliation(s)
- Marie-Charlotte Bourrienne
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Véronique Le Cam Duchez
- UNIROUEN, INSERM U1096, Normandie University, Rouen University Hospital, Hemostasis Unit and INSERM CIC-CRB 1404, Rouen, France
| | - Dorothée Faille
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Carine Farkh
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Juliette Gay
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Stéphane Loyau
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
| | - Clément Journé
- Université Paris Cité, INSERM UMS34, Fédération de Recherche en Imagerie Multimodalités, Faculté de Médecine X. Bichat, Paris, France
| | - Sébastien Dupont
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Véronique Ollivier
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Jean-Luc Villeval
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Valérie Edmond
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Martine Jandrot-Perrus
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
| | - Sylvie Labrouche-Colomer
- Université de Bordeaux, INSERM U1034, Biologie des maladies cardio-vasculaires & Laboratoire d’hématologie, Centre Hospitalier Universitaire de Bordeaux, Pessac, France
| | - Bruno Cassinat
- Laboratoire de Biologie Cellulaire, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Emmanuelle Verger
- Laboratoire de Biologie Cellulaire, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jean-Philippe Desilles
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Neuroradiologie interventionnelle, Hôpital Fondation Rothschild, Paris, France
| | - Benoît Ho-Tin-Noé
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Aude Triquenot Bagan
- Department of Neurology and INSERM CIC-CRB 1404, Rouen University Hospital, Rouen, France
| | - Mikaël Mazighi
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Neurologie, AP-HP, Hôpital Lariboisière, FHU NeuroVasc, Paris, France
| | - Nadine Ajzenberg
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
5
|
Haage TR, Charakopoulos E, Bhuria V, Baldauf CK, Korthals M, Handschuh J, Müller P, Li J, Harit K, Nishanth G, Frey S, Böttcher M, Fischer KD, Dudeck J, Dudeck A, Lipka DB, Schraven B, Green AR, Müller AJ, Mougiakakos D, Fischer T. Neutrophil-specific expression of JAK2-V617F or CALRmut induces distinct inflammatory profiles in myeloproliferative neoplasia. J Hematol Oncol 2024; 17:43. [PMID: 38853260 PMCID: PMC11163796 DOI: 10.1186/s13045-024-01562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Neutrophils play a crucial role in inflammation and in the increased thrombotic risk in myeloproliferative neoplasms (MPNs). We have investigated how neutrophil-specific expression of JAK2-V617F or CALRdel re-programs the functions of neutrophils. METHODS Ly6G-Cre JAK2-V617F and Ly6G-Cre CALRdel mice were generated. MPN parameters as blood counts, splenomegaly and bone marrow histology were compared to wild-type mice. Megakaryocyte differentiation was investigated using lineage-negative bone marrow cells upon in vitro incubation with TPO/IL-1β. Cytokine concentrations in serum of mice were determined by Mouse Cytokine Array. IL-1α expression in various hematopoietic cell populations was determined by intracellular FACS analysis. RNA-seq to analyse gene expression of inflammatory cytokines was performed in isolated neutrophils from JAK2-V617F and CALR-mutated mice and patients. Bioenergetics of neutrophils were recorded on a Seahorse extracellular flux analyzer. Cell motility of neutrophils was monitored in vitro (time lapse microscopy), and in vivo (two-photon microscopy) upon creating an inflammatory environment. Cell adhesion to integrins, E-selectin and P-selection was investigated in-vitro. Statistical analysis was carried out using GraphPad Prism. Data are shown as mean ± SEM. Unpaired, two-tailed t-tests were applied. RESULTS Strikingly, neutrophil-specific expression of JAK2-V617F, but not CALRdel, was sufficient to induce pro-inflammatory cytokines including IL-1 in serum of mice. RNA-seq analysis in neutrophils from JAK2-V617F mice and patients revealed a distinct inflammatory chemokine signature which was not expressed in CALR-mutant neutrophils. In addition, IL-1 response genes were significantly enriched in neutrophils of JAK2-V617F patients as compared to CALR-mutant patients. Thus, JAK2-V617F positive neutrophils, but not CALR-mutant neutrophils, are pathogenic drivers of inflammation in MPN. In line with this, expression of JAK2-V617F or CALRdel elicited a significant difference in the metabolic phenotype of neutrophils, suggesting a stronger inflammatory activity of JAK2-V617F cells. Furthermore, JAK2-V617F, but not CALRdel, induced a VLA4 integrin-mediated adhesive phenotype in neutrophils. This resulted in reduced neutrophil migration in vitro and in an inflamed vessel. This mechanism may contribute to the increased thrombotic risk of JAK2-V617F patients compared to CALR-mutant individuals. CONCLUSIONS Taken together, our findings highlight genotype-specific differences in MPN-neutrophils that have implications for the differential pathophysiology of JAK2-V617F versus CALR-mutant disease.
Collapse
Affiliation(s)
- Tobias Ronny Haage
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Emmanouil Charakopoulos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Vikas Bhuria
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Conny K Baldauf
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mark Korthals
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Handschuh
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Müller
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Juan Li
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Kunjan Harit
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Gopala Nishanth
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stephanie Frey
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Martin Böttcher
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel B Lipka
- Section of Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Anthony R Green
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Andreas J Müller
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Thomas Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany.
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Liu W, Pircher J, Schuermans A, Ul Ain Q, Zhang Z, Honigberg MC, Yalcinkaya M, Nakao T, Pournamadri A, Xiao T, Hajebrahimi MA, Wasner L, Stegner D, Petzold T, Natarajan P, Massberg S, Tall AR, Schulz C, Wang N. Jak2 V617F clonal hematopoiesis promotes arterial thrombosis via platelet activation and cross talk. Blood 2024; 143:1539-1550. [PMID: 38142422 PMCID: PMC11033586 DOI: 10.1182/blood.2023022260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/21/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023] Open
Abstract
ABSTRACT JAK2 V617F (JAK2VF) clonal hematopoiesis (CH) has been associated with atherothrombotic cardiovascular disease (CVD). We assessed the impact of Jak2VF CH on arterial thrombosis and explored the underlying mechanisms. A meta-analysis of 3 large cohort studies confirmed the association of JAK2VF with CVD and with platelet counts and adjusted mean platelet volume (MPV). In mice, 20% or 1.5% Jak2VF CH accelerated arterial thrombosis and increased platelet activation. Megakaryocytes in Jak2VF CH showed elevated proplatelet formation and release, increasing prothrombogenic reticulated platelet counts. Gp1ba-Cre-mediated expression of Jak2VF in platelets (VFGp1ba) increased platelet counts to a similar level as in 20% Jak2VF CH mice while having no effect on leukocyte counts. Like Jak2VF CH mice, VFGp1ba mice showed enhanced platelet activation and accelerated arterial thrombosis. In Jak2VF CH, both Jak2VF and wild-type (WT) platelets showed increased activation, suggesting cross talk between mutant and WT platelets. Jak2VF platelets showed twofold to threefold upregulation of COX-1 and COX-2, particularly in young platelets, with elevated cPLA2 activation and thromboxane A2 production. Compared with controls, conditioned media from activated Jak2VF platelets induced greater activation of WT platelets that was reversed by a thromboxane receptor antagonist. Low-dose aspirin ameliorated carotid artery thrombosis in VFGp1ba and Jak2VF CH mice but not in WT control mice. This study shows accelerated arterial thrombosis and platelet activation in Jak2VF CH with a major role of increased reticulated Jak2VF platelets, which mediate thromboxane cross talk with WT platelets and suggests a potential beneficial effect of aspirin in JAK2VF CH.
Collapse
Affiliation(s)
- Wenli Liu
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Joachim Pircher
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Art Schuermans
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Faculty of Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Qurrat Ul Ain
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhe Zhang
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael C. Honigberg
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Cardiology Division, Brigham and Women’s Hospital, Boston, MA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Ashley Pournamadri
- Biomedical Informatics Graduate Training Program, Stanford University, Stanford, CA
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Mohammad Ali Hajebrahimi
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Lisa Wasner
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - David Stegner
- Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Tobias Petzold
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Steffen Massberg
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Christian Schulz
- Department of Cardiology, Medical Clinic I, University Hospital, Ludwig Maximilian University, Munich, Germany
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
7
|
He F, Laranjeira AB, Kong T, Lin S, Ashworth KJ, Liu A, Lasky NM, Fisher DA, Cox MJ, Fulbright MC, Antunes-Heck L, Yu L, Brakhane M, Gao B, Sykes SM, D’Alessandro A, Di Paola J, Oh ST. Multiomic profiling reveals metabolic alterations mediating aberrant platelet activity and inflammation in myeloproliferative neoplasms. J Clin Invest 2024; 134:e172256. [PMID: 38060311 PMCID: PMC10836808 DOI: 10.1172/jci172256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/06/2023] [Indexed: 02/02/2024] Open
Abstract
Platelets from patients with myeloproliferative neoplasms (MPNs) exhibit a hyperreactive phenotype. Here, we found elevated P-selectin exposure and platelet-leukocyte aggregates indicating activation of platelets from essential thrombocythemia (ET) patients. Single-cell RNA-seq analysis of primary samples revealed significant enrichment of transcripts related to platelet activation, mTOR, and oxidative phosphorylation in ET patient platelets. These observations were validated via proteomic profiling. Platelet metabolomics revealed distinct metabolic phenotypes consisting of elevated ATP generation accompanied by increases in the levels of multiple intermediates of the tricarboxylic acid cycle, but lower α-ketoglutarate (α-KG) in MPN patients. Inhibition of PI3K/AKT/mTOR signaling significantly reduced metabolic responses and hyperreactivity in MPN patient platelets, while α-KG supplementation markedly reduced oxygen consumption and ATP generation. Ex vivo incubation of platelets from both MPN patients and Jak2 V617F-knockin mice with α-KG supplementation significantly reduced platelet activation responses. Oral α-KG supplementation of Jak2 V617F mice decreased splenomegaly and reduced hematocrit, monocyte, and platelet counts. Finally, α-KG treatment significantly decreased proinflammatory cytokine secretion from MPN CD14+ monocytes. Our results reveal a previously unrecognized metabolic disorder in conjunction with aberrant PI3K/AKT/mTOR signaling that contributes to platelet hyperreactivity in MPN patients.
Collapse
Affiliation(s)
- Fan He
- Division of Hematology, Department of Medicine, and
| | | | - Tim Kong
- Division of Hematology, Department of Medicine, and
| | - Shuyang Lin
- Division of Hematology, Department of Medicine, and
| | - Katrina J. Ashworth
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alice Liu
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nina M. Lasky
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | - Lilian Antunes-Heck
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - LaYow Yu
- Division of Hematology, Department of Medicine, and
| | | | - Bei Gao
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephen M. Sykes
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jorge Di Paola
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephen T. Oh
- Division of Hematology, Department of Medicine, and
- Immunomonitoring Laboratory, Center for Human Immunology and Immunotherapy Programs, and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Guy A, Garcia G, Gourdou-Latyszenok V, Wolff-Trombini L, Josserand L, Kimmerlin Q, Favre S, Kilani B, Marty C, Boulaftali Y, Labrouche-Colomer S, Mansier O, James C. Platelets and neutrophils cooperate to induce increased neutrophil extracellular trap formation in JAK2V617F myeloproliferative neoplasms. J Thromb Haemost 2024; 22:172-187. [PMID: 37678548 DOI: 10.1016/j.jtha.2023.08.028] [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: 02/28/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Neutrophils participate in the pathogenesis of thrombosis through the formation of neutrophil extracellular traps (NETs). Thrombosis is the main cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). Recent studies have shown an increase in NET formation (NETosis) both in patients with JAK2V617F neutrophils and in mouse models, and reported the participation of NETosis in the pathophysiology of thrombosis in mice. OBJECTIVES This study investigated whether JAK2V617F neutrophils are sufficient to promote thrombosis or whether their cooperation with other blood cell types is necessary. METHODS NETosis was studied in PF4iCre;Jak2V617F/WT mice expressing JAK2V617F in all hematopoietic lineages, as occurs in MPNs, and in MRP8Cre;Jak2V617F/WT mice in which JAK2V617F is expressed only in leukocytes. RESULTS In PF4iCre;Jak2V617F/WT mice, an increase in NETosis and spontaneous lung thrombosis abrogated by DNAse administration were observed. The absence of spontaneous NETosis or lung thrombosis in MRP8Cre;Jak2V617F/WT mice suggested that mutated neutrophils alone are not sufficient to induce thrombosis. Ex vivo experiments demonstrated that JAK2V617F-mutated platelets trigger NETosis by JAK2V617F-mutated neutrophils. Aspirin treatment in PF4iCre;Jak2V617F/WT mice reduced NETosis and reduced lung thrombosis. In cytoreductive-therapy-free patients with MPN treated with aspirin, plasma NET marker concentrations were lower than that in patients with MPN not treated with aspirin. CONCLUSION Our study demonstrates that JAK2V617F neutrophils alone are not sufficient to promote thrombosis; rather, platelets cooperate with neutrophils to promote NETosis in vivo. A new role for aspirin in thrombosis prevention in MPNs was also identified.
Collapse
Affiliation(s)
- Alexandre Guy
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France. https://twitter.com/Alexandreguy6
| | - Geoffrey Garcia
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/GeofGarciaVirginie
| | - Virginie Gourdou-Latyszenok
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/GourdouV
| | - Laura Wolff-Trombini
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/TrombiniWolff
| | - Lara Josserand
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Quentin Kimmerlin
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Simon Favre
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Badr Kilani
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Caroline Marty
- Institut national de la santé et de la recherche médicale, UMR1287, University of Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Yacine Boulaftali
- Paris Diderot University, Institut national de la santé et de la recherche médicale, Unité Mixte de Recherche_S1148, Laboratory for Vascular Translational Science, Paris, France
| | - Sylvie Labrouche-Colomer
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France
| | - Olivier Mansier
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France
| | - Chloé James
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France.
| |
Collapse
|
9
|
Jones E, Dillon B, Swan D, Thachil J. Practical management of the haemorrhagic complications of myeloproliferative neoplasms. Br J Haematol 2022; 199:313-321. [PMID: 35724983 PMCID: PMC9796684 DOI: 10.1111/bjh.18322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023]
Abstract
Myeloproliferative neoplasms can be associated with bleeding manifestations which can cause significant morbidities. Although haematologists are aware of the likelihood of this complication in the setting of myeloproliferative neoplasms, it may often be overlooked especially in patients with no extreme elevation of blood counts and those with myelofibrosis. Acquired von Willebrand syndrome and platelet dysfunction are the two common diagnoses to be considered in this regard. In this review article, we discuss the mechanisms for the development of these rare bleeding disorders, their diagnosis and practical management.
Collapse
Affiliation(s)
- Edward Jones
- Department of HaematologySt James' HospitalDublinIreland
| | - Bryan Dillon
- Department of HaematologySt James' HospitalDublinIreland
| | - Dawn Swan
- National University IrelandGalwayIreland
| | - Jecko Thachil
- Department of HaematologyManchester University Hospitals NHS Foundation TrustManchesterUK
| |
Collapse
|
10
|
Murphy AJ, Dragoljevic D, Natarajan P, Wang N. Hematopoiesis of Indeterminate Potential and Atherothrombotic Risk. Thromb Haemost 2022; 122:1435-1442. [PMID: 35445383 PMCID: PMC9420552 DOI: 10.1055/a-1830-2147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/23/2022] [Indexed: 11/28/2022]
Abstract
Hematopoiesis is the process of blood production, essential for the continued supply of immune cells and red blood cells. However, the proliferative nature of hematopoietic stem cells (HSCs) renders them susceptible to developing somatic mutations. HSCs carrying a mutation can gain a selective advantage over normal HSCs and result in hematological disorders. One such disorder is termed clonal hematopoiesis of indeterminate potential (CHIP), a premalignant state associated with aging, where the mutant HSCs are responsible for producing a small portion of mature immune cells in the circulation and subsequently in tissues. People with CHIP have been shown to have an increased risk of mortality due to cardiovascular disease (CVD). Why this occurs is under rigorous investigation, but the majority of the studies to date have suggested that increased atherosclerosis is due to heightened inflammatory cytokine release from mutant lesional macrophages. However, given CHIP is driven by several mutations, other hematopoietic lineages can be altered to promote CVD. In this review we explore the relationship between mutations in genes causing CHIP and atherothrombotic disorders, along with potential mechanisms of enhanced clonal outgrowth and potential therapies and strategies to slow CHIP progression.
Collapse
Grants
- National Heart, Lung, and Blood Institute R01HL148071
- National Health and Medical Research Council APP1194329
- National Heart, Lung, and Blood Institute R01HL142711
- National Heart, Lung, and Blood Institute R01HL148050
- National Heart, Lung, and Blood Institute R01HL151283
- National Heart, Lung, and Blood Institute R01HL127564
- National Institute of Diabetes and Digestive and Kidney Diseases R01DK125782
- National Heart, Lung, and Blood Institute R01HL118567
- Fondation Leducq TNE-18CVD04
- National Heart, Lung, and Blood Institute R01HL135242
- National Heart, Lung, and Blood Institute R01HL151152
- R01 HL148050 NHLBI NIH HHS
- National Heart, Lung, and Blood Institute R01HL148565
- National Health and Medical Research Council APP1142938
Collapse
Affiliation(s)
- Andrew J. Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Dragana Dragoljevic
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Pradeep Natarajan
- Cardiology Division, Department of Medicine, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University Medical Center, New York, United States
| |
Collapse
|
11
|
Clonal hematopoiesis and cardiovascular disease in cancer patients and survivors. Thromb Res 2022; 213 Suppl 1:S107-S112. [DOI: 10.1016/j.thromres.2021.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/22/2022]
|
12
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022; 23:ijms23063206. [PMID: 35328626 PMCID: PMC8954909 DOI: 10.3390/ijms23063206] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K. Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-67-15338; Fax: +49-391-67-15852
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
13
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022. [PMID: 35328626 DOI: 10.3390/ijms23063206.pmid:35328626;pmcid:pmc8954909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
14
|
Carobbio A, Vannucchi AM, De Stefano V, Masciulli A, Guglielmelli P, Loscocco GG, Ramundo F, Rossi E, Kanthi Y, Tefferi A, Barbui T. Neutrophil-to-lymphocyte ratio is a novel predictor of venous thrombosis in polycythemia vera. Blood Cancer J 2022; 12:28. [PMID: 35145055 PMCID: PMC8831521 DOI: 10.1038/s41408-022-00625-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 11/09/2022] Open
Abstract
We investigated the neutrophil-to-lymphocyte ratio (NLR) as a predictor of thrombosis in polycythemia vera (PV). After a median follow-up of 2.51 years, of 1508 PV patients enrolled in the ECLAP study, 82 and 84 developed arterial and venous thrombosis, respectively. Absolute counts of total leukocytes, neutrophils, lymphocytes, platelets, and the NLR were tested by generalized additive models (GAM) to evaluate their trend in continuous scale of thrombotic risk. Only for venous thrombosis, we showed that baseline absolute neutrophil and lymphocyte counts were on average respectively higher (median: 6.8 × 109/L, p = 0.002) and lower (median: 1.4 × 109/L, p = 0.001), leading to increased NLR values (median: 5.1, p = 0.002). In multivariate analysis, the risk of venous thrombosis was independently associated with previous venous events (HR = 5.48, p ≤ 0.001) and NLR values ≥5 (HR = 2.13, p = 0.001). Moreover, the relative risk in both low- and high-standard risk groups was almost doubled in the presence of NLR ≥ 5. These findings were validated in two Italian independent external cohorts (Florence, n = 282 and Rome, n = 175) of contemporary PV patients. Our data support recent experimental work that venous thrombosis is controlled by innate immune cells and highlight that NLR is an inexpensive and easily accessible prognostic biomarker of venous thrombosis.
Collapse
Affiliation(s)
| | - Alessandro Maria Vannucchi
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Valerio De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Arianna Masciulli
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Paola Guglielmelli
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Giuseppe Gaetano Loscocco
- Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Department of Experimental and Clinical Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Francesco Ramundo
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Elena Rossi
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | | | - Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tiziano Barbui
- FROM Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy.
| |
Collapse
|
15
|
Papageorgiou L, Elalamy I, Vandreden P, Gerotziafas GT. Thrombotic and Hemorrhagic Issues Associated with Myeloproliferative Neoplasms. Clin Appl Thromb Hemost 2022; 28:10760296221097969. [PMID: 35733370 PMCID: PMC9234921 DOI: 10.1177/10760296221097969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Thrombotic and hemorrhagic complications are related to a significant rate of
morbidity and mortality in patients with myeloproliferative neoplasms (MPNs),
they are therefore called “thrombohemorrhagic” syndromes. Several clinical
factors, such as age and presence of cardiovascular comorbidities are
responsible for thrombotic complications. High blood counts, platelet
alterations, presence of JAK2 mutation and possibly of other CHIP mutations such
as TET2, DNMT3A, and ASXL1, procoagulant microparticles, NETs formation,
endothelial activation and neo-angiogenesis are some of the parameters
accounting for hypercoagulability in patients with myeloproliferative neoplasms.
Bleeding complications emerge as a result of platelet exhaustion. They can be
also linked to a functional deficiency of von Willebrand factor, when platelet
counts rise above 1000G/L. The mainstay of management consists on preventing
hemostatic complications, by antiplatelet and/or anticoagulant treatment and
myelosuppressive agents in high-risk patients.Circumstances related to a high
thrombohemorrhagic risk, such as pregnancy and the perioperative period, prompt
for specific management with regards to anticoagulation and myelosuppression
treatment type. In order to apply a patient-specific treatment strategy, there
is a need for a risk score assessment tool encompassing clinical parameters and
hemostasis biomarkers.
Collapse
Affiliation(s)
- Loula Papageorgiou
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France
| | - Ismail Elalamy
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France.,The First I.M. Sechenov Moscow State Medical University, Moscow, Russia
| | - Patrick Vandreden
- Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France.,Clinical Research Department, Diagnostica Stago, Gennevilliers, France
| | - Grigoris T Gerotziafas
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France
| |
Collapse
|
16
|
Bleeding diathesis in mice lacking JAK2 in platelets. Blood Adv 2021; 5:2969-2981. [PMID: 34342643 DOI: 10.1182/bloodadvances.2020003032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine kinase JAK2 is a critical component of intracellular JAK/STAT cytokine signaling cascades that is prevalent in hematopoietic cells, such as hematopoietic stem cells and megakaryocytes (MKs). Individuals expressing the somatic JAK2 V617F mutation commonly develop myeloproliferative neoplasms (MPNs) associated with venous and arterial thrombosis, a leading cause of mortality. The role of JAK2 in hemostasis remains unclear. We investigated the role of JAK2 in platelet hemostatic function using Jak2fl/fl Pf4-Cre (Jak2Plt-/-) mice lacking JAK2 in platelets and MKs. Jak2Plt-/- mice developed MK hyperplasia and splenomegaly associated with severe thrombocytosis and bleeding. This notion was supported by failure to occlude in a ferric chloride carotid artery injury model and by a cremaster muscle laser-induced injury assay, in which Jak2Plt-/- platelets failed to form stable thrombi. Jak2Plt-/- platelets formed thrombi poorly after adhesion to type 1 collagen under arterial shear rates. Jak2Plt-/- platelets spread poorly on collagen under static conditions or on fibrinogen in response to the collagen receptor GPVI-specific agonist, collagen-related peptide (CRP). After activation with collagen, CRP, or the CLEC-2 agonist rhodocytin, Jak2Plt-/- platelets displayed decreased α-granule secretion and integrin αIIbβ3 activation or aggregation, but showed normal responses to thrombin. Jak2Plt-/- platelets had impaired intracellular signaling when activated via GPVI, as assessed by tyrosine phosphorylation. Together, the results show that JAK2 deletion impairs platelet immunoreceptor tyrosine-based activation motif signaling and hemostatic function in mice and suggest that aberrant JAK2 signaling in patients with MPNs affects GPVI signaling, leading to hemostatic platelet function.
Collapse
|
17
|
Pharmacological Inhibition of Insulin Growth Factor-1 Receptor (IGF-1R) Alone or in Combination With Ruxolitinib Shows Therapeutic Efficacy in Preclinical Myeloproliferative Neoplasm Models. Hemasphere 2021; 5:e565. [PMID: 33954282 PMCID: PMC8092367 DOI: 10.1097/hs9.0000000000000565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/22/2021] [Indexed: 11/26/2022] Open
Abstract
Even after development of the JAK1/JAK2 inhibitor ruxolitinib, myeloproliferative neoplasm (MPN) patients require novel therapeutic options. While ruxolitinib can considerably improve quality of life and prolong survival, it does not modify the natural disease course in most patients. Moreover, resistance develops with prolonged use. Therefore, various combination treatments are currently being investigated. Published data provide a compelling rationale for the inhibition of insulin growth factor-1 receptor (IGF-1R) signaling in MPN. Here we report that genetic and pharmacological inhibition of IGF-1R selectively reduced Jak2V617F-driven cytokine-independent proliferation ex vivo. Two different structurally unrelated IGF-1R inhibitors ameliorated disease phenotype in a murine MPN model and significantly prolonged survival. Moreover, in mice, low-dose ruxolitinib synergized with IGF-1R inhibition to increase survival. Our data demonstrate preclinical efficacy of IGF-1R inhibition in a murine MPN model.
Collapse
|
18
|
The pathobiology of thrombosis, microvascular disease, and hemorrhage in the myeloproliferative neoplasms. Blood 2021; 137:2152-2160. [PMID: 33649757 DOI: 10.1182/blood.2020008109] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Thrombotic, vascular, and bleeding complications are the most common causes of morbidity and mortality in the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). In these disorders, circulating red cells, leukocytes, and platelets, as well as some vascular endothelial cells, each have abnormalities that are cell-intrinsic to the MPN driver mutations they harbor (eg, JAK2 V617F). When these cells are activated in the MPNs, their interactions with each other create a highly proadhesive and prothrombotic milieu in the circulation that predisposes patients with MPN to venous, arterial, and microvascular thrombosis and occlusive disease. Bleeding problems in the MPNs are caused by the MPN blood cell-initiated development of acquired von Willebrand disease. The inflammatory state created by MPN stem cells in their microenvironment extends systemically to amplify the clinical thrombotic tendency and, at the same time, preferentially promote further MPN stem cell clonal expansion, thereby generating a vicious cycle that favors a prothrombotic state in these diseases.
Collapse
|
19
|
Reeves BN, Beckman JD. Novel Pathophysiological Mechanisms of Thrombosis in Myeloproliferative Neoplasms. Curr Hematol Malig Rep 2021; 16:304-313. [PMID: 33876389 DOI: 10.1007/s11899-021-00630-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Thrombosis remains a leading cause of morbidity and mortality in BCR/ABL negative myeloproliferative neoplasms (MPN). Circulating blood cells are both increased in quantity and qualitatively abnormal in MPN, resulting in an increased thrombotic risk. Herein, we review recently elucidated mechanisms of MPN thrombosis and discuss implications of drugs currently under investigation for MPN. RECENT FINDINGS Recent studies highlight that in JAK2V617F granulocytes and platelets, thrombo-inflammatory genes are upregulated. Furthermore, in JAK2V617F granulocytes, protein expression of integrin CD11b, tissue factor, and leukocyte alkaline phosphatase are all increased. Overall, myeloid cells, namely neutrophils, may contribute in several ways, such as through increased adhesion via β1 integrin binding to VCAM1, increased infiltration, and enhanced inducibility to extrude neutrophil extracellular traps. Non-myeloid inflammatory cells may also contribute via secretion of cytokines. With regard to red blood cells, number, rigidity, adhesion, and generation of microvesicles may lead to increased vascular resistance as well as increased cell-cell interactions that promote rolling and adhesion. Platelets may also contribute in a similar fashion. Lastly, the vasculature is also increasingly appreciated, as several studies have demonstrated increased endothelial expression of pro-coagulant and pro-adhesive proteins, such as von Willebrand factor or P-selectin in JAK2V617F endothelial cells. With the advent of molecular diagnostics, MPN therapeutics are advancing beyond cytoreduction. Our increased understanding of pro-inflammatory and thrombotic pathophysiology in MPN provides a rational basis for evaluation of in-development MPN therapeutics to reduce thrombosis.
Collapse
Affiliation(s)
- Brandi N Reeves
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan D Beckman
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, 420 Delaware St. SE, MMC 480, Minneapolis, MN, 55455, USA.
| |
Collapse
|
20
|
N-acetylcysteine inhibits thrombosis in a murine model of myeloproliferative neoplasm. Blood Adv 2021; 4:312-321. [PMID: 31978215 DOI: 10.1182/bloodadvances.2019000967] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Thrombosis is a major cause of mortality in patients with myeloproliferative neoplasms (MPNs), though there is currently little to offer patients with MPN beyond aspirin and cytoreductive therapies such as hydroxyurea for primary prevention. Thrombogenesis in MPN involves multiple cellular mechanisms, including platelet activation and neutrophil-extracellular trap formation; therefore, an antithrombotic agent that targets one or more of these processes would be of therapeutic benefit in MPN. Here, we treated the JAK2V617F knockin mouse model of polycythemia vera with N-acetylcysteine (NAC), a sulfhydryl-containing compound with broad effects on glutathione replenishment, free radical scavenging, and reducing disulfide bonds, to investigate its antithrombotic effects in the context of MPN. Strikingly, NAC treatment extended the lifespan of JAK2V617F mice without impacting blood counts or splenomegaly. Using an acute pulmonary thrombosis model in vivo, we found that NAC reduced thrombus formation to a similar extent as the irreversible platelet inhibitor aspirin. In vitro analysis of platelet activation revealed that NAC reduced thrombin-induced platelet-leukocyte aggregate formation in JAK2V617F mice. Furthermore, NAC reduced neutrophil extracellular trap formation in primary human neutrophils from patients with MPN as well as healthy controls. These results provide evidence that N-acetylcysteine inhibits thrombosis in JAK2V617F mice and provide a pre-clinical rationale for investigating NAC as a therapeutic to reduce thrombotic risk in MPN.
Collapse
|
21
|
Benlabiod C, Dagher T, Marty C, Villeval JL. Lessons from mouse models of MPN. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 366:125-185. [PMID: 35153003 DOI: 10.1016/bs.ircmb.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the past decades, a variety of MPN mouse models have been developed to express in HSC the main mutations identified in patients: JAK2V617F, CALRdel52 or ins5 and MPLW515L. These models mimic quite faithfully human PV or ET with their natural evolutions into MF and their hemostasis complications, demonstrating the driver function of these mutations in MPN. Here, we review these models and show how they have improved our general understanding of MPN regarding (1) the mechanisms of fibrosis, thrombosis/hemorrhages and disease initiation, (2) the roles of additional mutations and signaling pathways in disease progression and (3) the preclinical development of novel therapies. We also address controversial results between these models and remind how these models may differ from human MPN onset and also how basically mice are not humans, encouraging caution when one draw lessons from mice to humans. Furthermore, the contribution of germline genetic predisposition, HSC and niche aging, metabolic, oxidative, replicative or genotoxic stress, inflammation, immune escape and additional mutations need to be considered in further investigations to encompass the full complexity of human MPN in mice.
Collapse
Affiliation(s)
- Camelia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Tracy Dagher
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
22
|
Guy A, Poisson J, James C. Pathogenesis of cardiovascular events in BCR-ABL1-negative myeloproliferative neoplasms. Leukemia 2021; 35:935-955. [PMID: 33658660 DOI: 10.1038/s41375-021-01170-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
Thrombosis, both in arterial and venous territories, is the major complication of myeloproliferative neoplasms and is responsible for a high rate of morbidity and mortality. The currently accepted risk factors are an age over 60 years and a history of thrombosis. However, many complex mechanisms contribute to this increased prothrombotic risk, with involvement of all blood cell types, plasmatic factors, and endothelial cells. Besides, some cardiovascular events may originate from arterial vasospasm that could contribute to thrombotic complications. In this review, we discuss recent results obtained in mouse models in the light of data obtained from clinical studies. We emphasize on actors of thrombosis that are currently not targeted with current therapeutics but could be promising targets, i.e, neutrophil extracellular traps and vascular reactivity.
Collapse
Affiliation(s)
- Alexandre Guy
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Johanne Poisson
- Inserm, Centre de recherche sur l'inflammation, University of Paris, Paris, France.,Geriatrics Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Chloe James
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France. .,Laboratoire d'Hématologie, CHU de Bordeaux, Pessac, France.
| |
Collapse
|
23
|
Murine Modeling of Myeloproliferative Neoplasms. Hematol Oncol Clin North Am 2021; 35:253-265. [PMID: 33641867 DOI: 10.1016/j.hoc.2020.11.007] [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: 11/20/2022]
Abstract
Myeloproliferative neoplasms, such as polycythemia vera, essential thrombocythemia, and primary myelofibrosis, are bone marrow disorders that result in the overproduction of mature clonal myeloid elements. Identification of recurrent genetic mutations has been described and aid in diagnosis and prognostic determination. Mouse models of these mutations have confirmed the biologic significance of these mutations in myeloproliferative neoplasm disease biology and provided greater insights on the pathways that are dysregulated with each mutation. The models are useful tools that have led to preclinical testing and provided data as validation for future myeloproliferative neoplasm clinical trials.
Collapse
|
24
|
Poisson J, Tanguy M, Davy H, Camara F, El Mdawar MB, Kheloufi M, Dagher T, Devue C, Lasselin J, Plessier A, Merchant S, Blanc-Brude O, Souyri M, Mougenot N, Dingli F, Loew D, Hatem SN, James C, Villeval JL, Boulanger CM, Rautou PE. Erythrocyte-derived microvesicles induce arterial spasms in JAK2V617F myeloproliferative neoplasm. J Clin Invest 2021; 130:2630-2643. [PMID: 32045382 PMCID: PMC7190923 DOI: 10.1172/jci124566] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/04/2020] [Indexed: 12/16/2022] Open
Abstract
Arterial cardiovascular events are the leading cause of death in patients with JAK2V617F myeloproliferative neoplasms (MPNs). However, their mechanisms are poorly understood. The high prevalence of myocardial infarction without significant coronary stenosis or atherosclerosis in patients with MPNs suggests that vascular function is altered. The consequences of JAK2V617F mutation on vascular reactivity are unknown. We observe here increased responses to vasoconstrictors in arteries from Jak2V617F mice resulting from a disturbed endothelial NO pathway and increased endothelial oxidative stress. This response was reproduced in WT mice by circulating microvesicles isolated from patients carrying JAK2V617F and by erythrocyte-derived microvesicles from transgenic mice. Microvesicles of other cellular origins had no effect. This effect was observed ex vivo on isolated aortas, but also in vivo on femoral arteries. Proteomic analysis of microvesicles derived from JAK2V617F erythrocytes identified increased expression of myeloperoxidase as the likely mechanism accounting for their effect. Myeloperoxidase inhibition in microvesicles derived from JAK2V617F erythrocytes suppressed their effect on oxidative stress. Antioxidants such as simvastatin and N-acetyl cysteine improved arterial dysfunction in Jak2V617F mice. In conclusion, JAK2V617F MPNs are characterized by exacerbated vasoconstrictor responses resulting from increased endothelial oxidative stress caused by circulating erythrocyte-derived microvesicles. Simvastatin appears to be a promising therapeutic strategy in this setting.
Collapse
Affiliation(s)
- Johanne Poisson
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France.,Centre de recherche sur l'inflammation, Inserm, Université de Paris, Paris, France.,Geriatrics Department, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Marion Tanguy
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France.,Centre de recherche sur l'inflammation, Inserm, Université de Paris, Paris, France
| | - Hortense Davy
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Fatoumata Camara
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Marie-Belle El Mdawar
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Marouane Kheloufi
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Tracy Dagher
- Inserm U1170, Institut Gustave Roussy, Université Paris XI, Villejuif, France
| | - Cécile Devue
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Juliette Lasselin
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Aurélie Plessier
- Service d'Hépatologie, Pôle des Maladies de l'Appareil Digestif, Hôpital Beaujon, Département Hospitalo-Universitaire (DHU Unity), AP-HP, Clichy, France.,Centre de Référence des Maladies Vasculaires du Foie, French Network for Rare Liver Diseases (FILFOIE), European Reference Network (ERN), Clichy, France
| | - Salma Merchant
- Inserm U1170, Institut Gustave Roussy, Université Paris XI, Villejuif, France
| | - Olivier Blanc-Brude
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Michèle Souyri
- Inserm UMR S1131, University Hospital Institute (IHU), Université de Paris, Paris, France
| | - Nathalie Mougenot
- Inserm UMS 28, Phénotypage du petit animal, Plateforme d'expérimentations coeur-muscle-vaisseaux (PECMV), Sorbonne University, Paris, France
| | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Université de recherche PSL, Paris, France
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Université de recherche PSL, Paris, France
| | - Stephane N Hatem
- Inserm, UMR 1166, Institut de cardiométabolisme et nutrition (ICAN), Sorbonne University, Paris, France
| | - Chloé James
- Inserm U1034, Biology of Cardiovascular, Pessac, France.,University of Bordeaux, Pessac, France.,Laboratory of Hematology, Bordeaux University Hospital Center, Pessac, France
| | - Jean-Luc Villeval
- Inserm U1170, Institut Gustave Roussy, Université Paris XI, Villejuif, France
| | - Chantal M Boulanger
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France
| | - Pierre-Emmanuel Rautou
- Paris-Centre de recherche cardiovasculaire (PARCC), Université de Paris, Paris, France.,Centre de recherche sur l'inflammation, Inserm, Université de Paris, Paris, France.,Inserm U1170, Institut Gustave Roussy, Université Paris XI, Villejuif, France.,Service d'Hépatologie, Pôle des Maladies de l'Appareil Digestif, Hôpital Beaujon, Département Hospitalo-Universitaire (DHU Unity), AP-HP, Clichy, France
| |
Collapse
|
25
|
Stanly TA, Suman R, Rani GF, O’Toole PJ, Kaye PM, Hitchcock IS. Quantitative Optical Diffraction Tomography Imaging of Mouse Platelets. Front Physiol 2020; 11:568087. [PMID: 33041864 PMCID: PMC7526686 DOI: 10.3389/fphys.2020.568087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/25/2020] [Indexed: 11/13/2022] Open
Abstract
Platelets are specialized anucleate cells that play a major role in hemostasis following vessel injury. More recently, platelets have also been implicated in innate immunity and inflammation by directly interacting with immune cells and releasing proinflammatory signals. It is likely therefore that in certain pathologies, such as chronic parasitic infections and myeloid malignancies, platelets can act as mediators for hemostatic and proinflammatory responses. Fortunately, murine platelet function ex vivo is highly analogous to human, providing a robust model for functional comparison. However, traditional methods of studying platelet phenotype, function and activation status often rely on using large numbers of whole isolated platelet populations, which severely limits the number and type of assays that can be performed with mouse blood. Here, using cutting edge 3D quantitative phase imaging, holotomography, that uses optical diffraction tomography (ODT), we were able to identify and quantify differences in single unlabeled, live platelets with minimal experimental interference. We analyzed platelets directly isolated from whole blood of mice with either a JAK2V617F-positive myeloproliferative neoplasm (MPN) or Leishmania donovani infection. Image analysis of the platelets indicates previously uncharacterized differences in platelet morphology, including altered cell volume and sphericity, as well as changes in biophysical parameters such as refractive index (RI) and dry mass. Together, these data indicate that, by using holotomography, we were able to identify clear disparities in activation status and potential functional ability in disease states compared to control at the level of single platelets.
Collapse
Affiliation(s)
- Tess A. Stanly
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Rakesh Suman
- Technology Facility, Department of Biology, University of York, York, United Kingdom
| | - Gulab Fatima Rani
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Peter J. O’Toole
- Technology Facility, Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Ian S. Hitchcock
- York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
26
|
Matsuura S, Thompson CR, Belghasem ME, Bekendam RH, Piasecki A, Leiva O, Ray A, Italiano J, Yang M, Merill-Skoloff G, Chitalia VC, Flaumenhaft R, Ravid K. Platelet Dysfunction and Thrombosis in JAK2 V617F-Mutated Primary Myelofibrotic Mice. Arterioscler Thromb Vasc Biol 2020; 40:e262-e272. [PMID: 32814440 DOI: 10.1161/atvbaha.120.314760] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The risk of thrombosis in myeloproliferative neoplasms, such as primary myelofibrosis varies depending on the type of key driving mutation (JAK2 [janus kinase 2], CALR [calreticulin], and MPL [myeloproliferative leukemia protein or thrombopoietin receptor]) and the accompanying mutations in other genes. In the current study, we sought to examine the propensity for thrombosis, as well as platelet activation properties in a mouse model of primary myelofibrosis induced by JAK2V617F (janus kinase 2 with valine to phenylalanine substitution on codon 617) mutation. Approach and Results: Vav1-hJAK2V617F transgenic mice show hallmarks of primary myelofibrosis, including significant megakaryocytosis and bone marrow fibrosis, with a moderate increase in red blood cells and platelet number. This mouse model was used to study responses to 2 models of vascular injury and to investigate platelet properties. Platelets derived from the mutated mice have reduced aggregation in response to collagen, reduced thrombus formation and thrombus size, as demonstrated using laser-induced or FeCl3-induced vascular injury models, and increased bleeding time. Strikingly, the mutated platelets had a significantly reduced number of dense granules, which could explain impaired ADP secretion upon platelet activation, and a diminished second wave of activation. CONCLUSIONS Together, our study highlights for the first time the influence of a hyperactive JAK2 on platelet activation-induced ADP secretion and dense granule homeostasis, with consequent effects on platelet activation properties.
Collapse
Affiliation(s)
- Shinobu Matsuura
- Department of Medicine and Whitaker Cardiovascular Institute (S.M., C.R.T., A.P., O.L., K.R.), Boston University School of Medicine, MA
| | - Cristal R Thompson
- Department of Medicine and Whitaker Cardiovascular Institute (S.M., C.R.T., A.P., O.L., K.R.), Boston University School of Medicine, MA
| | | | - Roelof H Bekendam
- Department of Medicine (R.H.B.), Boston University School of Medicine, MA
| | - Andrew Piasecki
- Department of Medicine and Whitaker Cardiovascular Institute (S.M., C.R.T., A.P., O.L., K.R.), Boston University School of Medicine, MA
| | - Orly Leiva
- Department of Medicine and Whitaker Cardiovascular Institute (S.M., C.R.T., A.P., O.L., K.R.), Boston University School of Medicine, MA
| | - Anjana Ray
- Department of Medicine, Brigham and Women's Hospital, Boston MA (A.R., J.I.)
| | - Joseph Italiano
- Department of Medicine, Brigham and Women's Hospital, Boston MA (A.R., J.I.)
| | - Moua Yang
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.Y., G.M.-S., R.F.)
| | - Glenn Merill-Skoloff
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.Y., G.M.-S., R.F.)
| | - Vipul C Chitalia
- Renal Section, Department of Medicine (V.C.C.), Boston University School of Medicine, MA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.Y., G.M.-S., R.F.)
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute (S.M., C.R.T., A.P., O.L., K.R.), Boston University School of Medicine, MA
| |
Collapse
|
27
|
Description of a knock-in mouse model of JAK2V617F MPN emerging from a minority of mutated hematopoietic stem cells. Blood 2020; 134:2383-2387. [PMID: 31697834 DOI: 10.1182/blood.2019001163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/01/2019] [Indexed: 12/27/2022] Open
Abstract
The major weakness of most knock-in JAK2V617F mouse models is the presence of the JAK2 mutation in all rather than in a few hematopoietic stem cells (HSC), such as in human "early-stage" myeloproliferative neoplasms (MPN). Understanding the mechanisms of disease initiation is critical as underscored by the incidence of clonal hematopoiesis of indeterminate potential associated with JAK2V617F. Currently, such studies require competitive transplantation. Here, we report a mouse model obtained by crossing JAK2V617F/WT knock-in mice with PF4iCre transgenic mice. As expected, PF4iCre;JAK2V617F/WT mice developed an early thrombocytosis resulting from the expression of JAK2V617F in the megakaryocytes. However, these mice then developed a polycythemia vera-like phenotype at 10 weeks of age. Using mT/mG reporter mice, we demonstrated that Cre recombination was present in all hematopoietic compartments, including in a low number of HSC. The frequency of mutated cells increased along hematopoietic differentiation mimicking the clonal expansion observed in essential thrombocythemia and polycythemia vera patients. This model thus mimics the HSC compartment observed in early-stage MPN, with a small number of JAK2V617F HSC competing with a majority of JAK2WT HSC. PF4iCre;JAK2V617F/WT mice are a promising tool to investigate the mechanisms that regulate clonal dominance and progression to myelofibrosis.
Collapse
|
28
|
Liu YC, Illar GM, Bailey NG. Clinicopathologic characterisation of myeloid neoplasms with concurrent spliceosome mutations and myeloproliferative-neoplasm-associated mutations. J Clin Pathol 2020; 73:728-736. [PMID: 32217616 DOI: 10.1136/jclinpath-2020-206495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/29/2022]
Abstract
AIMS Spliceosome genes (SF3B1, SRSF2, U2AF1 and ZRSR2) are commonly mutated in myeloid neoplasms, particularly in myelodysplastic syndromes (MDS). JAK2, MPL and CALR mutations are associated with myeloproliferative neoplasms (MPN). Although SF3B1 and MPN-associated mutations frequently co-occur in the rare entity MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T), myeloid neoplasms with concurrent spliceosome and MPN-associated mutations encompass many disease entities and are not well characterised. METHODS Specimens from 2016 to 2019 with concurrent spliceosome and MPN-associated mutations were identified, and the clinicopathologic features were assessed. RESULTS The 36 cases were divided into mutational categories based on their spliceosome mutation. At diagnosis, cases with concurrent U2AF1 and MPN-associated mutations had lower leucocyte counts and platelet counts than did the other groups. Cases with mutant SRSF2 were more likely to have ASXL1 and IDH2 mutations, while U2AF1-mutated neoplasms were more likely to have an abnormal karyotype. The most common SF3B1 K700 and U2AF1 S34 mutational hotspots were underrepresented in our cohort of myeloid neoplasms with concurrent spliceosome and MPN-associated mutations, as SF3B1 and U2AF1 mutations tended to involve other codons. Numerous WHO-defined disease entities were represented in each spliceosome gene category; although MDS/MPN-RS-T were only identified in the group with SF3B1 mutations, they constituted only 1/4 of the neoplasms in the category. CONCLUSIONS Myeloid neoplasms with different mutant splicing factor and concurrent MPN-associated mutations demonstrate somewhat different clinical and pathologic features, but t he association between genotypes and phenotypes in these overlapping neoplasms is not straightforward.
Collapse
Affiliation(s)
- Yen-Chun Liu
- Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
29
|
Mornet C, Galinat H, Mingant F, Ianotto JC, Lippert E. [Thrombosis and platelet dysfunction in myeloproliferative neoplasms]. Rev Med Interne 2020; 41:319-324. [PMID: 32008800 DOI: 10.1016/j.revmed.2019.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Myeloproliferative neoplasms are acquired hematological malignancies, mainly affecting the adult and whose morbidity and mortality stems from haemostasis disorders. The most frequently encountered complications include thrombosis, affecting preferentially the arterial territory, but also atypical locations such as splanchnic vein thrombosis. The pathophysiology of these thromboses is complex and involves different actors: blood cells, endothelium and flow conditions. Numerous studies have been conducted to identify risk factors for thrombosis. To date, only two risk factors have been validated through prospective studies (age over 60 years old, history of thrombotic events) and allow classification of patients as "low risk" and "high risk" as the basis for current treatment recommendations. Haemorrhagic manifestations, less frequent than thrombosis, are mainly related to an alteration of primary haemostasis and are therefore manifested by mucocutaneous bleeding. In these patients, platelet dysfunctions and/or acquired Willebrand syndromes can be found. The pathophysiology of thrombosis and platelet dysfunction during myeloproliferative neoplasms remains to date partially unknown. In this review, we offer to focus on physiopathological mechanisms as well as the latest advances in their understanding.
Collapse
Affiliation(s)
- C Mornet
- Hématologie biologique, CHU de Brest, Brest, France
| | - H Galinat
- Hématologie biologique, CHU de Brest, Brest, France
| | - F Mingant
- Hématologie biologique, CHU de Brest, Brest, France
| | - J C Ianotto
- Hématologie clinique et thérapie cellulaire, CHU de Brest, Brest, France
| | - E Lippert
- Hématologie biologique, CHU de Brest, Brest, France; Inserm, EFS, UMR 1078, GGB, Université Brest, Brest, France.
| |
Collapse
|
30
|
Wolach O, Sellar RS, Martinod K, Cherpokova D, McConkey M, Chappell RJ, Silver AJ, Adams D, Castellano CA, Schneider RK, Padera RF, DeAngelo DJ, Wadleigh M, Steensma DP, Galinsky I, Stone RM, Genovese G, McCarroll SA, Iliadou B, Hultman C, Neuberg D, Mullally A, Wagner DD, Ebert BL. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci Transl Med 2019; 10:10/436/eaan8292. [PMID: 29643232 DOI: 10.1126/scitranslmed.aan8292] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 01/09/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022]
Abstract
Thrombosis is a major cause of morbidity and mortality in Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), clonal disorders of hematopoiesis characterized by activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. Neutrophil extracellular trap (NET) formation, a component of innate immunity, has been linked to thrombosis. We demonstrate that neutrophils from patients with MPNs are primed for NET formation, an effect blunted by pharmacological inhibition of JAK signaling. Mice with conditional knock-in of Jak2V617F, the most common molecular driver of MPN, have an increased propensity for NET formation and thrombosis. Inhibition of JAK-STAT signaling with the clinically available JAK2 inhibitor ruxolitinib abrogated NET formation and reduced thrombosis in a deep vein stenosis murine model. We further show that expression of PAD4, a protein required for NET formation, is increased in JAK2V617F-expressing neutrophils and that PAD4 is required for Jak2V617F-driven NET formation and thrombosis in vivo. Finally, in a population study of more than 10,000 individuals without a known myeloid disorder, JAK2V617F-positive clonal hematopoiesis was associated with an increased incidence of thrombosis. In aggregate, our results link JAK2V617F expression to NET formation and thrombosis and suggest that JAK2 inhibition may reduce thrombosis in MPNs through cell-intrinsic effects on neutrophil function.
Collapse
Affiliation(s)
- Ofir Wolach
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA.,Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 49100, Israel
| | - Rob S Sellar
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA.,Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Kimberly Martinod
- Program in Cellular and Molecular Medicine and Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine and Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Marie McConkey
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ryan J Chappell
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexander J Silver
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dylan Adams
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Rebekka K Schneider
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA.,Department of Hematology, Cancer Institute, Erasmus Medical Center, Rotterdam 2040, Netherlands
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Martha Wadleigh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - David P Steensma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Ilene Galinsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Giulio Genovese
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven A McCarroll
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Bozenna Iliadou
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 76, Sweden
| | - Christina Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm SE-171 76, Sweden
| | - Donna Neuberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Ann Mullally
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA.,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine and Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin L Ebert
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA. .,Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
31
|
Guy A, James C. [Pathogenesis of thrombosis in JAK2V617F myeloproliferative neoplasms]. Med Sci (Paris) 2019; 35:651-658. [PMID: 31532377 DOI: 10.1051/medsci/2019133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BCR-ABL negative myeloproliferative neoplasms are acquired hematologic diseases characterized by blood cell proliferation and that include polycythemia vera (PV), essential thrombocytemia (ET) and primary myelofibrosis (PMF). Occurring of venous and arterial thrombosis is the main complication of these diseases. Risk factors for thrombosis are individuals older than 60 and history of thrombosis. The mechanisms leading to thrombosis are complex and involve several blood compartments, plasmatic factors and endothelial cells. Over the last years, new actors of thrombosis have been discovered.
Collapse
Affiliation(s)
- Alexandre Guy
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| | - Chloé James
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| |
Collapse
|
32
|
Perner F, Perner C, Ernst T, Heidel FH. Roles of JAK2 in Aging, Inflammation, Hematopoiesis and Malignant Transformation. Cells 2019; 8:cells8080854. [PMID: 31398915 PMCID: PMC6721738 DOI: 10.3390/cells8080854] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/22/2022] Open
Abstract
Clonal alterations in hematopoietic cells occur during aging and are often associated with the establishment of a subclinical inflammatory environment. Several age-related conditions and diseases may be initiated or promoted by these alterations. JAK2 mutations are among the most frequently mutated genes in blood cells during aging. The most common mutation within the JAK2 gene is JAK2-V617F that leads to constitutive activation of the kinase and thereby aberrant engagement of downstream signaling pathways. JAK2 mutations can act as central drivers of myeloproliferative neoplasia, a pre-leukemic and age-related malignancy. Likewise, hyperactive JAK-signaling is a hallmark of immune diseases and critically influences inflammation, coagulation and thrombosis. In this review we aim to summarize the current knowledge on JAK2 in clonal hematopoiesis during aging, the role of JAK-signaling in inflammation and lymphocyte biology and JAK2 function in age-related diseases and malignant transformation.
Collapse
Affiliation(s)
- Florian Perner
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany
- Dana-Farber Cancer Institute, Department of Pediatric Oncology, Harvard University, Boston, MA 02467, USA
| | - Caroline Perner
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, 02129 MA, USA
| | - Thomas Ernst
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany
| | - Florian H Heidel
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany.
- Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany.
| |
Collapse
|
33
|
Leiva O, Bekendam RH, Garcia BD, Thompson C, Cantor A, Chitalia V, Ravid K. Emerging Factors Implicated in Fibrotic Organ-Associated Thrombosis: The Case of Two Organs. TH OPEN 2019; 3:e165-e170. [PMID: 31259299 PMCID: PMC6598088 DOI: 10.1055/s-0039-1692204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022] Open
Abstract
Thrombosis is at the heart of cardiovascular complications observed in specific diseases. A heightened thrombosis risk above that in general population in diseases such as myelofibrosis and chronic kidney disease implicates disease-specific mediators of thrombosis. This relative lack of information regarding the mechanisms of thrombosis in specific organ pathologies hitherto has remained limited. Evolving literature implicates some soluble factors in the blood of patients with discrete disorders, inflicting fundamental changes in the components of thrombosis. In this era of precision medicine, integrating these disease-specific factors in a comprehensive thrombotic risk assessment of patients is imperative in guiding therapeutic decisions. A complex network of mechanisms regulates each organ pathology and resultant thrombotic phenotypes. This review surveys different effectors of thrombogenicity associated with two pathologically fibrotic organs used as model systems, the bone marrow and kidney, as well as focuses attention to a common inducer of fibrosis and thrombosis, lysyl oxidase.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Roelof H Bekendam
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Brenda D Garcia
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Department of Medicine, Mount Auburn Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Cristal Thompson
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Alan Cantor
- Children's Hospital Boston, Boston, Massachusetts, United States
| | - Vipul Chitalia
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States.,VA Boston Healthcare System, Boston, Massachusetts, United States
| | - Katya Ravid
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
34
|
Gordeuk VR, Key NS, Prchal JT. Re-evaluation of hematocrit as a determinant of thrombotic risk in erythrocytosis. Haematologica 2019; 104:653-658. [PMID: 30872370 PMCID: PMC6442963 DOI: 10.3324/haematol.2018.210732] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Here we critically evaluate the role of elevated hematocrit as the principal determinant of thrombotic risk in polycythemia and erythrocytosis, defined by an expansion of red cell mass. Since red cell volume determination is no longer readily available, in clinical practice, polycythemia and erythrocytosis are defined by elevated hemoglobin and hematocrit. Thrombosis is common in Chuvash erythrocytosis and polycythemia vera. Although the increased thrombotic risk is assumed to be due to the elevated hematocrit and an associated increase in blood viscosity, thrombosis does not accompany most types of erythrocytosis. We review studies indicating that the occurrence of thrombosis in Chuvash erythrocytosis is independent of hematocrit, that the thrombotic risk is paradoxically increased by phlebotomy in Chuvash erythrocytosis, and that, when compared to chemotherapy, phlebotomy is associated with increased thrombotic risk in polycythemia vera. Inherited and environmental causes that lead to polycythemia and erythrocytosis are accompanied by diverse cellular changes that could directly affect thrombotic risk, irrespective of the elevated hematocrit. The pressing issue in these disorders is to define factors other than elevated hematocrit that determine thrombotic risk. Defining these predisposing factors in polycythemia and erythrocytosis should then lead to rational therapies and facilitate development of targeted interventions.
Collapse
Affiliation(s)
- Victor R Gordeuk
- Division of Hematology and Oncology, University of Illinois at Chicago, IL
| | - Nigel S Key
- Division of Hematology-Oncology and UNC Hemophilia and Thrombosis Center, UNC, Chapel Hill, NC
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, University of Utah and Huntsman Cancer Center, Salt Lake City, UT, USA
| |
Collapse
|
35
|
WEISEL JW, LITVINOV RI. Red blood cells: the forgotten player in hemostasis and thrombosis. J Thromb Haemost 2019; 17:271-282. [PMID: 30618125 PMCID: PMC6932746 DOI: 10.1111/jth.14360] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Indexed: 12/14/2022]
Abstract
New evidence has stirred up a long-standing but undeservedly forgotten interest in the role of erythrocytes, or red blood cells (RBCs), in blood clotting and its disorders. This review summarizes the most recent research that describes the involvement of RBCs in hemostasis and thrombosis. There are both quantitative and qualitative changes in RBCs that affect bleeding and thrombosis, as well as interactions of RBCs with cellular and molecular components of the hemostatic system. The changes in RBCs that affect hemostasis and thrombosis include RBC counts or hematocrit (modulating blood rheology through viscosity) and qualitative changes, such as deformability, aggregation, expression of adhesive proteins and phosphatidylserine, release of extracellular microvesicles, and hemolysis. The pathogenic mechanisms implicated in thrombotic and hemorrhagic risk include variable adherence of RBCs to the vessel wall, which depends on the functional state of RBCs and/or endothelium, modulation of platelet reactivity and platelet margination, alterations of fibrin structure and reduced susceptibility to fibrinolysis, modulation of nitric oxide availability, and the levels of von Willebrand factor and factor VIII in blood related to the ABO blood group system. RBCs are involved in platelet-driven contraction of clots and thrombi that results in formation of a tightly packed array of polyhedral erythrocytes, or polyhedrocytes, which comprises a nearly impermeable barrier that is important for hemostasis and wound healing. The revisited notion of the importance of RBCs is largely based on clinical and experimental associations between RBCs and thrombosis or bleeding, implying that RBCs are a prospective therapeutic target in hemostatic and thrombotic disorders.
Collapse
Affiliation(s)
- J. W. WEISEL
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - R. I. LITVINOV
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
36
|
Edelmann B, Gupta N, Schnoeder TM, Oelschlegel AM, Shahzad K, Goldschmidt J, Philipsen L, Weinert S, Ghosh A, Saalfeld FC, Nimmagadda SC, Müller P, Braun-Dullaeus R, Mohr J, Wolleschak D, Kliche S, Amthauer H, Heidel FH, Schraven B, Isermann B, Müller AJ, Fischer T. JAK2-V617F promotes venous thrombosis through β1/β2 integrin activation. J Clin Invest 2018; 128:4359-4371. [PMID: 30024857 DOI: 10.1172/jci90312] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
JAK2-V617F-positive chronic myeloproliferative neoplasia (CMN) commonly displays dysfunction of integrins and adhesion molecules expressed on platelets, erythrocytes, and leukocytes. However, the mechanism by which the 2 major leukocyte integrin chains, β1 and β2, may contribute to CMN pathophysiology remained unclear. β1 (α4β1; VLA-4) and β2 (αLβ2; LFA-1) integrins are essential regulators for attachment of leukocytes to endothelial cells. We here showed enhanced adhesion of granulocytes from mice with JAK2-V617F knockin (JAK2+/VF mice) to vascular cell adhesion molecule 1- (VCAM1-) and intercellular adhesion molecule 1-coated (ICAM1-coated) surfaces. Soluble VCAM1 and ICAM1 ligand binding assays revealed increased affinity of β1 and β2 integrins for their respective ligands. For β1 integrins, this correlated with a structural change from the low- to the high-affinity conformation induced by JAK2-V617F. JAK2-V617F triggered constitutive activation of the integrin inside-out signaling molecule Rap1, resulting in translocation toward the cell membrane. Employing a venous thrombosis model, we demonstrated that neutralizing anti-VLA-4 and anti-β2 integrin antibodies suppress pathologic thrombosis as observed in JAK2+/VF mice. In addition, aberrant homing of JAK2+/VF leukocytes to the spleen was inhibited by neutralizing anti-β2 antibodies and by pharmacologic inhibition of Rap1. Thus, our findings identified cross-talk between JAK2-V617F and integrin activation promoting pathologic thrombosis and abnormal trafficking of leukocytes to the spleen.
Collapse
Affiliation(s)
- Bärbel Edelmann
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Nibedita Gupta
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Tina M Schnoeder
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Internal Medicine II, Hematology and Oncology, University Hospital Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Anja M Oelschlegel
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Institute of Anatomy
| | | | | | - Lars Philipsen
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, and
| | - Soenke Weinert
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Department of Cardiology and Angiology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Aniket Ghosh
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Felix C Saalfeld
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Subbaiah Chary Nimmagadda
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Müller
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Rüdiger Braun-Dullaeus
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Department of Cardiology and Angiology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Mohr
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, and
| | - Denise Wolleschak
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| | - Stefanie Kliche
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, and
| | - Holger Amthauer
- Department of Radiology and Nuclear Medicine, University Hospital, Magdeburg, Germany
| | - Florian H Heidel
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Internal Medicine II, Hematology and Oncology, University Hospital Jena, Jena, Germany.,Leibniz Institute on Aging, Fritz-Lipmann-Institute, Jena, Germany
| | - Burkhart Schraven
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, and.,Helmholtz Centre for Infection Research, Department of Immune Control, Braunschweig, Germany
| | - Berend Isermann
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Clinical Chemistry and Pathobiochemistry
| | - Andreas J Müller
- Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Institute of Molecular and Clinical Immunology, and.,Helmholtz Centre for Infection Research, Department of Immune Control, Braunschweig, Germany
| | - Thomas Fischer
- Department of Hematology and Oncology, Medical Center, Otto-von-Guericke University, Magdeburg, Germany.,Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
37
|
Poisson J, Hilscher MB, Tanguy M, Hammoutene A, Boulanger CM, Villeval JL, Simonetto DA, Valla D, Shah VH, Rautou PE. Endothelial JAK2 V617F does not enhance liver lesions in mice with Budd-Chiari syndrome. J Hepatol 2018; 68:1086-1087. [PMID: 29360553 DOI: 10.1016/j.jhep.2018.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/05/2018] [Accepted: 01/09/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Johanne Poisson
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Moira B Hilscher
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Marion Tanguy
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Adel Hammoutene
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Chantal M Boulanger
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Luc Villeval
- INSERM, Institut Gustave Roussy, INSERM U1170, Villejuif, France; University Paris XI, Villejuif, France
| | - Douglas A Simonetto
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Dominique Valla
- Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France
| | - Vijay H Shah
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Pierre-Emmanuel Rautou
- INSERM, UMR-970, Paris Cardiovascular Research Center - PARCC, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Service d'hépatologie, DHU Unity Hôpital Beaujon, APHP, Clichy, France; Université Denis Diderot-Paris 7, Sorbonne Paris Cité, 75018 Paris, France.
| |
Collapse
|
38
|
Chen CC, Chiu CC, Lee KD, Hsu CC, Chen HC, Huang THM, Hsiao SH, Leu YW. JAK2V617F influences epigenomic changes in myeloproliferative neoplasms. Biochem Biophys Res Commun 2017; 494:470-476. [PMID: 29066347 DOI: 10.1016/j.bbrc.2017.10.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022]
Abstract
Negative valine (V) to phenylalanine (F) switch at the Janus kinase (JAK2) 617 codon (V617F) is the dominant driver mutation in patients with myeloproliferative neoplasms (MPNs). JAK2V617F was proved to be sufficient for cell transformation; however, independent mutations might influence the following epigenomic modifications. To assess the JAK2V617F-induced downstream epigenomic changes without interferences, we profiled the epigenomic changes in ectopically expressed JAK2V617F in Ba/F3 cells. Antibodies against phosphorylated signal transducer and activator of transcription 3 (pSTAT3) and enhancer of zeste homolog 2 (EZH2) were used for chromatin-immunoprecipitation sequencing (ChIP-seq) to detect the downstream epigenomic targets in the JAK2-STAT3 signaling pathway. To confirm the JAK2V617F-induced epigenetic changes in vivo, DNA methylation changes in the target loci in patients with MPNs were detected through methylation-specific polymerase chain reaction and were clustered against the changes within controls. We found that ectopically expressed JAK2V617F in Ba/F3 cells reduced the binding specificity; it was associated with cis-regulatory elements and recognized DNA motifs in both pSTAT3-downstream and EZH2-associated targets. Overlapping target loci between the control and JAK2V617F were <3% and 0.4%, respectively, as identified through pSTAT3 and EZH2 ChIP-seq. Furthermore, the methylation changes in the direct target loci (FOXH1, HOXC9, and SRF) were clustered independently from the control locus (L1TD1) and other mutation genes (HMGA2 and Lin28A) in the analyzed MPN samples. Therefore, JAK2V617F influences target binding in both pSTAT3 and EZH2. Without mutations in epigenetic regulators, JAK2V617F can induce downstream epigenomic modifications. Thus, epigenetic changes in JAK2 downstream targets might be trackable in vivo.
Collapse
Affiliation(s)
- Chih-Cheng Chen
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Chang Gung University College of Medicine, Taoyuan, Taiwan; Chang Gung Institute of Technology, Taoyuan, Taiwan
| | - Chia-Chen Chiu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Chang Gung University College of Medicine, Taoyuan, Taiwan; Chang Gung Institute of Technology, Taoyuan, Taiwan; Human Epigenomics Center, Department of Life Science, Institute of Molecular Biology and Institute of Biomedical Science, National Chung Cheng University, Chia-Yi, Taiwan
| | - Kuan-Der Lee
- Division of Hematology and Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chia-Chen Hsu
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi, Chang Gung University College of Medicine, Taoyuan, Taiwan; Chang Gung Institute of Technology, Taoyuan, Taiwan
| | - Hong-Chi Chen
- Department of Life Science and Gene Therapy Division, Tzu-Chi University and Hospital, Hualien, Taiwan
| | - Tim H-M Huang
- Cancer Therapy and Research Center, Department of Molecular Medicine and Institute of Biotechnology, School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shu-Huei Hsiao
- Human Epigenomics Center, Department of Life Science, Institute of Molecular Biology and Institute of Biomedical Science, National Chung Cheng University, Chia-Yi, Taiwan.
| | - Yu-Wei Leu
- Human Epigenomics Center, Department of Life Science, Institute of Molecular Biology and Institute of Biomedical Science, National Chung Cheng University, Chia-Yi, Taiwan.
| |
Collapse
|
39
|
Zhao B, Mei Y, Cao L, Zhang J, Sumagin R, Yang J, Gao J, Schipma MJ, Wang Y, Thorsheim C, Zhao L, Stalker T, Stein B, Wen QJ, Crispino JD, Abrams CS, Ji P. Loss of pleckstrin-2 reverts lethality and vascular occlusions in JAK2V617F-positive myeloproliferative neoplasms. J Clin Invest 2017; 128:125-140. [PMID: 29202466 DOI: 10.1172/jci94518] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/17/2017] [Indexed: 12/19/2022] Open
Abstract
V617F driver mutation of JAK2 is the leading cause of the Philadelphia-chromosome-negative myeloproliferative neoplasms (MPNs). Although thrombosis is a leading cause of mortality and morbidity in MPNs, the mechanisms underlying their pathogenesis are unclear. Here, we identified pleckstrin-2 (Plek2) as a downstream target of the JAK2/STAT5 pathway in erythroid and myeloid cells, and showed that it is upregulated in a JAK2V617F-positive MPN mouse model and in patients with MPNs. Loss of Plek2 ameliorated JAK2V617F-induced myeloproliferative phenotypes including erythrocytosis, neutrophilia, thrombocytosis, and splenomegaly, thereby reverting the widespread vascular occlusions and lethality in JAK2V617F-knockin mice. Additionally, we demonstrated that a reduction in red blood cell mass was the main contributing factor in the reversion of vascular occlusions. Thus, our study identifies Plek2 as an effector of the JAK2/STAT5 pathway and a key factor in the pathogenesis of JAK2V617F-induced MPNs, pointing to Plek2 as a viable target for the treatment of MPNs.
Collapse
Affiliation(s)
- Baobing Zhao
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Yang Mei
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Lan Cao
- Department of Pathology, Feinberg School of Medicine, and.,Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Jingxin Zhang
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Jing Yang
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Juehua Gao
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Matthew J Schipma
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yanfeng Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chelsea Thorsheim
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Liang Zhao
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy Stalker
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brady Stein
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.,Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Qiang Jeremy Wen
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.,Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John D Crispino
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.,Division of Hematology and Oncology, Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Charles S Abrams
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, and.,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
40
|
Abstract
Red blood cells (RBCs) have historically been considered passive bystanders in thrombosis. However, clinical and epidemiological studies have associated quantitative and qualitative abnormalities in RBCs, including altered hematocrit, sickle cell disease, thalassemia, hemolytic anemias, and malaria, with both arterial and venous thrombosis. A growing body of mechanistic studies suggests that RBCs can promote thrombus formation and enhance thrombus stability. These findings suggest that RBCs may contribute to thrombosis pathophysiology and reveal potential strategies for therapeutically targeting RBCs to reduce thrombosis.
Collapse
|
41
|
Abstract
In contrast to congenital platelet disorders, which are rare, acquired platelet dysfunctions are more common in clinical practice. Their main causes are medications and systemic/hematologic diseases. Typical clinical manifestations are mucosal bleeding, epistaxis, or superficial epidermal bleeding normally of modest entity. In most cases, the molecular mechanisms underlying impaired platelet function are not fully established, making it difficult to optimize patient care. We here provide a short overview of the various forms of acquired platelet disorders, with a particular focus on recent mechanistic studies on platelet dysfunction in von Willebrand disease.
Collapse
Affiliation(s)
- Caterina Casari
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wolfgang Bergmeier
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
42
|
Elevated hematocrit enhances platelet accumulation following vascular injury. Blood 2017; 129:2537-2546. [PMID: 28251913 DOI: 10.1182/blood-2016-10-746479] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/22/2017] [Indexed: 12/30/2022] Open
Abstract
Red blood cells (RBCs) demonstrate procoagulant properties in vitro, and elevated hematocrit is associated with reduced bleeding and increased thrombosis risk in humans. These observations suggest RBCs contribute to thrombus formation. However, effects of RBCs on thrombosis are difficult to assess because humans and mice with elevated hematocrit typically have coexisting pathologies. Using an experimental model of elevated hematocrit in healthy mice, we measured effects of hematocrit in 2 in vivo clot formation models. We also assessed thrombin generation, platelet-thrombus interactions, and platelet accumulation in thrombi ex vivo, in vitro, and in silico. Compared with controls, mice with elevated hematocrit (RBCHIGH) formed thrombi at a faster rate and had a shortened vessel occlusion time. Thrombi in control and RBCHIGH mice did not differ in size or fibrin content, and there was no difference in levels of circulating thrombin-antithrombin complexes. In vitro, increasing the hematocrit increased thrombin generation in the absence of platelets; however, this effect was reduced in the presence of platelets. In silico, direct numerical simulations of whole blood predicted elevated hematocrit increases the frequency and duration of interactions between platelets and a thrombus. When human whole blood was perfused over collagen at arterial shear rates, elevating the hematocrit increased the rate of platelet deposition and thrombus growth. These data suggest RBCs promote arterial thrombosis by enhancing platelet accumulation at the site of vessel injury. Maintaining a normal hematocrit may reduce arterial thrombosis risk in humans.
Collapse
|
43
|
How J, Zhou A, Oh ST. Splanchnic vein thrombosis in myeloproliferative neoplasms: pathophysiology and molecular mechanisms of disease. Ther Adv Hematol 2017; 8:107-118. [PMID: 28246554 PMCID: PMC5305004 DOI: 10.1177/2040620716680333] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are the most common underlying prothrombotic disorder found in patients with splanchnic vein thrombosis (SVT). Clinical risk factors for MPN-associated SVTs include younger age, female sex, concomitant hypercoagulable disorders, and the JAK2 V617F mutation. These risk factors are distinct from those associated with arterial or deep venous thrombosis (DVT) in MPN patients, suggesting disparate disease mechanisms. The pathophysiology of SVT is thought to derive from local interactions between activated blood cells and the unique splanchnic endothelial environment. Other mutations commonly found in MPNs, including CALR and MPL, are rare in MPN-associated SVT. The purpose of this article is to review the clinical and molecular risk factors for MPN-associated SVT, with particular focus on the possible mechanisms of SVT formation in MPN patients.
Collapse
Affiliation(s)
- Joan How
- Division of Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Amy Zhou
- Division of Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephen T. Oh
- Division of Hematology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8125, St Louis, MO 63110, USA
| |
Collapse
|
44
|
Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood 2016; 129:667-679. [PMID: 28028029 DOI: 10.1182/blood-2016-10-695940] [Citation(s) in RCA: 423] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/06/2016] [Indexed: 02/07/2023] Open
Abstract
The genetic landscape of classical myeloproliferative neoplasm (MPN) is in large part elucidated. The MPN-restricted driver mutations, including those in JAK2, calreticulin (CALR), and myeloproliferative leukemia virus (MPL), abnormally activate the cytokine receptor/JAK2 pathway and their downstream effectors, more particularly the STATs. The most frequent mutation, JAK2V617F, activates the 3 main myeloid cytokine receptors (erythropoietin receptor, granulocyte colony-stimulating factor receptor, and MPL) whereas CALR or MPL mutants are restricted to MPL activation. This explains why JAK2V617F is associated with polycythemia vera, essential thrombocythemia (ET), and primary myelofibrosis (PMF) whereas CALR and MPL mutants are found in ET and PMF. Other mutations in genes involved in epigenetic regulation, splicing, and signaling cooperate with the 3 MPN drivers and play a key role in the PMF pathogenesis. Mutations in epigenetic regulators TET2 and DNMT3A are involved in disease initiation and may precede the acquisition of JAK2V617F. Other mutations in epigenetic regulators such as EZH2 and ASXL1 also play a role in disease initiation and disease progression. Mutations in the splicing machinery are predominantly found in PMF and are implicated in the development of anemia or pancytopenia. Both heterogeneity of classical MPNs and prognosis are determined by a specific genomic landscape, that is, type of MPN driver mutations, association with other mutations, and their order of acquisition. However, factors other than somatic mutations play an important role in disease initiation as well as disease progression such as germ line predisposition, inflammation, and aging. Delineation of these environmental factors will be important to better understand the precise pathogenesis of MPN.
Collapse
|
45
|
Appelmann I, Kreher S, Parmentier S, Wolf HH, Bisping G, Kirschner M, Bergmann F, Schilling K, Brümmendorf TH, Petrides PE, Tiede A, Matzdorff A, Griesshammer M, Riess H, Koschmieder S. Diagnosis, prevention, and management of bleeding episodes in Philadelphia-negative myeloproliferative neoplasms: recommendations by the Hemostasis Working Party of the German Society of Hematology and Medical Oncology (DGHO) and the Society of Thrombosis and Hemostasis Research (GTH). Ann Hematol 2016; 95:707-18. [PMID: 26916570 DOI: 10.1007/s00277-016-2621-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 12/31/2022]
Abstract
Philadelphia-negative myeloproliferative neoplasms (Ph-negative MPN) comprise a heterogeneous group of chronic hematologic malignancies. The quality of life, morbidity, and mortality of patients with MPN are primarily affected by disease-related symptoms, thromboembolic and hemorrhagic complications, and progression to myelofibrosis and acute leukemia. Major bleeding represents a common and important complication in MPN, and the incidence of such bleeding events will become even more relevant in the future due to the increasing disease prevalence and survival of MPN patients. This review discusses the causes, differential diagnoses, prevention, and management of bleeding episodes in patients with MPN, aiming at defining updated standards of care in these often challenging situations.
Collapse
Affiliation(s)
- Iris Appelmann
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Stephan Kreher
- Department of Hematology and Oncology, Charite Berlin, Berlin, Germany
| | - Stefani Parmentier
- Department of Hematology, Oncology, and Palliative Medicine, Rems-Murr-Klinikum Winnenden, Winnenden, Germany
| | - Hans-Heinrich Wolf
- Department of Internal Medicine IV, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Guido Bisping
- Department of Medicine I, Mathias Spital Rheine, Rheine, Germany
| | - Martin Kirschner
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Frauke Bergmann
- Medizinisches Versorgungszentrum Wagnerstibbe, Hannover, Germany
| | - Kristina Schilling
- Department of Hematology and Oncology, University Hospital Jena, Jena, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Petro E Petrides
- Hematology Oncology Centre, Ludwig Maximilians University of Munich Medical School, Munich, Germany
| | - Andreas Tiede
- Department of Haematology, Haemostasis, Oncology and Stem-Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Axel Matzdorff
- Clinic for Internal Medicine II, Dept. of Hematology, Oncology, Asklepios Clinic Uckermark, Schwedt/Oder, Germany
| | | | - Hanno Riess
- Department of Hematology and Oncology, Charite Berlin, Berlin, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
46
|
Lysyl oxidase is associated with increased thrombosis and platelet reactivity. Blood 2016; 127:1493-501. [PMID: 26755713 DOI: 10.1182/blood-2015-02-629667] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 12/17/2015] [Indexed: 01/26/2023] Open
Abstract
Lysyl oxidase (LOX) is overexpressed in various pathologies associated with thrombosis, such as arterial stenosis and myeloproliferative neoplasms (MPNs). LOX is elevated in the megakaryocytic lineage of mouse models of MPNs and in patients with MPNs. To gain insight into the role of LOX in thrombosis and platelet function without compounding the influences of other pathologies, transgenic mice expressing LOX in wild-type megakaryocytes and platelets (Pf4-Lox(tg/tg)) were generated. Pf4-Lox(tg/tg) mice had a normal number of platelets; however, time to vessel occlusion after endothelial injury was significantly shorter in Pf4-Lox(tg/tg) mice, indicating a higher propensity for thrombus formation in vivo. Exploring underlying mechanisms, we found that Pf4-Lox(tg/tg) platelets adhere better to collagen and have greater aggregation response to lower doses of collagen compared with controls. Platelet activation in response to the ligand for collagen receptor glycoprotein VI (cross-linked collagen-related peptide) was unaffected. However, the higher affinity of Pf4-Lox(tg/tg) platelets to the collagen sequence GFOGER implies that the collagen receptor integrin α2β1 is affected by LOX. Taken together, our findings demonstrate that LOX enhances platelet activation and thrombosis.
Collapse
|
47
|
Bonnard T, Hagemeyer CE. Ferric Chloride-induced Thrombosis Mouse Model on Carotid Artery and Mesentery Vessel. J Vis Exp 2015:e52838. [PMID: 26167713 DOI: 10.3791/52838] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Severe thrombosis and its ischemic consequences such as myocardial infarction, pulmonary embolism and stroke are major worldwide health issues. The ferric chloride injury is now a well-established technique to rapidly and accurately induce the formation of thrombi in exposed veins or artery of small and large diameter. This model has played a key role in the study of the pathophysiology of thrombosis, in the discovery and validation of novel antithrombotic drugs and in the understanding of the mechanism of action of these new agents. Here, the implementation of this technique on a mesenteric vessel and carotid artery in mice is presented. The method describes how to label circulating leukocytes and platelets with a fluorescent dye and to observe, by intravital microscopy on the exposed mesentery, their accumulation at the injured vessel wall which leads to the formation of a thrombus. On the carotid artery, the occlusion caused by the clot formation is measured by monitoring the blood flow with a Doppler probe.
Collapse
Affiliation(s)
- Thomas Bonnard
- Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute
| | | |
Collapse
|
48
|
Strassel C, Kubovcakova L, Mangin PH, Ravanat C, Freund M, Skoda RC, Denis CV, Dupuis A, Herbrecht R, Gachet C, Lanza F. Haemorrhagic and thrombotic diatheses in mouse models with thrombocytosis. Thromb Haemost 2014; 113:414-25. [PMID: 25298269 DOI: 10.1160/th14-08-0667] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 09/01/2014] [Indexed: 12/18/2022]
Abstract
We studied haemostasis in two mouse models with thrombocytosis caused by different pathogenic mechanisms. In one strain (Yall;Mpl-/-) thrombocytosis is driven by a misbalance between thrombopoietin and its receptor, whereas in the other strain, thrombocytosis is caused by expressing a human JAK2-V617F transgene (FF1) that depends on activation by Cre-recombinase (VavCre;FF1, MxCre;FF1). Thrombotic responses were increased following some, but not all types of challenges. In a vaso-occlusive thrombotic model following collagen-adrenaline injection we found increased mortality in both strains. Arterial thrombosis, examined after ferric chloride-induced carotid injury, was accelerated but with little impact on maximal thrombus size. In a vena cava stasis model, clots were of similar size as in wild-type controls, but exhibited a different composition with a higher platelet to fibrin ratio. Both thrombocytosis strains displayed increased haemorrhagic tendency in a tail bleeding assay. Yall;Mpl and VavCre;FF1 displayed a lower proportion of the more reactive high-molecular-weight forms of von Willebrand factor in their plasma, mimicking essential thrombocythaemia with very high platelet counts. Bleeding could not be explained by clear defects in platelet activation, which were normal or only weakly decreased. In conclusion, these models of thrombocytosis recapitulate several features of the haemorrhagic and thrombotic diatheses in ET and PV demonstrating potentials but also some limitations to study these major complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - François Lanza
- François Lanza, UMR S_949 - EFS-Alsace, 10, rue Spielmann, 67065 Strasbourg Cedex, France, Tel.: +33 388 21 25 25, Fax: +33 388 21 25 21, E-mail:
| |
Collapse
|
49
|
Is JAK2V617F finally off the hook? Blood 2014; 124:992-3. [DOI: 10.1182/blood-2014-07-583856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|