89 results on '"Kellie R. Machlus"'
Search Results
2. Dynamic actin/septin network in megakaryocytes coordinates proplatelet elaboration
- Author
-
Isabelle C. Becker, Adrian R. Wilkie, Bret A. Unger, Anthony R. Sciaudone, Farheen Fatima, I-Ting Tsai, Ke Xu, Kellie R. Machlus, and Joseph E. Italiano
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Megakaryocytes (MK) undergo extensive cytoskeletal rearrangements as they give rise to platelets. While cortical microtubule sliding has been implicated in proplatelet formation, the role of the actin cytoskeleton in proplatelet elongation is less understood. It is assumed that actin filament reorganization is important for platelet generation given that mouse models with mutations in actin-associated proteins exhibit thrombocytopenia. However, due to the essential role of the actin network during MK development, a differential understanding of the contribution of the actin cytoskeleton on proplatelet release is lacking. Here, we reveal that inhibition of actin polymerization impairs the formation of elaborate proplatelets by hampering proplatelet extension and bead formation along the proplatelet shaft, which was mostly independent of changes in cortical microtubule sliding. We identify Cdc42 and its downstream effectors, septins, as critical regulators of intracellular actin dynamics in MK, inhibition of which, similarly to inhibition of actin polymerization, impairs proplatelet movement and beading. Super-resolution microscopy revealed a differential association of distinctive septins with the actin and microtubule cytoskeleton, respectively, which was disrupted upon septin inhibition and diminished intracellular filamentous actin dynamics. In vivo, septins, similarly to F-actin, were subject to changes in expression upon enforcing proplatelet formation through prior platelet depletion. In summary, we demonstrate that a Cdc42/septin axis is not only important for MK maturation and polarization, but is further required for intracellular actin dynamics during proplatelet formation.
- Published
- 2023
- Full Text
- View/download PDF
3. Fluorescence artifact correction in the thrombin generation assay: Necessity for correction algorithms in procoagulant samples
- Author
-
William C. Chang, Joseph W. Jackson, Kellie R. Machlus, Alisa S. Wolberg, and Mikhail V. Ovanesov
- Subjects
blood coagulation factors ,calibration ,hemostasis ,plasma ,thrombin ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Introduction The thrombin generation (TG) test is a global hemostasis assay sensitive to procoagulant conditions. However, some TG assays may underestimate elevated TG when the thrombin fluorogenic substrate is depleted or fluorescence is attenuated by the inner filter effect (IFE). Objectives We sought to elucidate the extent to which procoagulant conditions require correcting for fluorogenic substrate depletion and/or IFE. Methods We analyzed corrections for substrate depletion and IFE and their effect on TG parameters in plasma samples with elevated blood coagulation factors in the presence or absence of thrombomodulin via commercial calibrated automated thrombogram (CAT) platform and in‐house software capable of internal thrombin calibration with or without CAT‐like artifact correction. Results Elevated thrombin peak height (TPH) and endogenous thrombin potential (ETP) were detected with 2× and 4× increases in blood coagulation factors I, V, VIII, IX, X, and XI, or prothrombin in the presence or absence of artifact correction. The effect of the CAT algorithm was evident in TG curves from both low procoagulant (thrombomodulin‐supplemented) and procoagulant (factor‐supplemented) plasma samples. However, in all samples, with the exception of elevated prothrombin, CAT’s correction was small (
- Published
- 2021
- Full Text
- View/download PDF
4. Venous thromboembolism research priorities: A scientific statement from the American Heart Association and the International Society on Thrombosis and Haemostasis
- Author
-
Mary Cushman, Geoffrey D. Barnes, Mark A. Creager, Jose A. Diaz, Peter K. Henke, Kellie R. Machlus, Marvin T. Nieman, Alisa S. Wolberg, and the American Heart Association Council on Peripheral Vascular Disease, Council on Arteriosclerosis, Thrombosis, and Vascular Biology, Council on Cardiovascular and Stroke Nursing, Council on Clinical Cardiology, Council on Epidemiology and Prevention, and International Society on Thrombosis and Haemostasis (ISTH)
- Subjects
hemostasis ,postthrombotic syndrome ,pulmonary embolism ,thrombosis ,venous thromboembolism ,venous thrombosis ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Venous thromboembolism (VTE) is a major cause of morbidity and mortality. The impact of the Surgeon General’s Call to Action in 2008 has been lower than expected given the public health impact of this disease. This scientific statement highlights future research priorities in VTE, developed by experts and a crowdsourcing survey across 16 scientific organizations. At the fundamental research level (T0), researchers need to identify pathobiologic causative mechanisms for the 50% of patients with unprovoked VTE and better understand mechanisms that differentiate hemostasis from thrombosis. At the human level (T1), new methods for diagnosing, treating, and preventing VTE will allow tailoring of diagnostic and therapeutic approaches to individuals. At the patient level (T2), research efforts are required to understand how foundational evidence impacts care of patients (eg, biomarkers). New treatments, such as catheter‐based therapies, require further testing to identify which patients are most likely to experience benefit. At the practice level (T3), translating evidence into practice remains challenging. Areas of overuse and underuse will require evidence‐based tools to improve care delivery. At the community and population level (T4), public awareness campaigns need thorough impact assessment. Large population‐based cohort studies can elucidate the biologic and environmental underpinings of VTE and its complications. To achieve these goals, funding agencies and training programs must support a new generation of scientists and clinicians who work in multidisciplinary teams to solve the pressing public health problem of VTE.
- Published
- 2020
- Full Text
- View/download PDF
5. Blood and Bone: The quarantine chronicles
- Author
-
Kirk A. Taylor and Kellie R. Machlus
- Subjects
hematopoiesis ,hemostasis ,megakaryocyte ,platelet ,thrombosis ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract In the midst of the chaos of the global pandemic, the online daily webinar series Blood and Bone was created. The series started with a blank schedule on a google doc and, with enthusiasm and participation from the hematopoiesis and hemostasis/thrombosis communities, was quickly filled through September 2020. In the absence of any editing of the speaker list, a diverse, well‐balanced, and scientifically exciting program emerged. The seminar is hosted on Zoom and live‐streamed on YouTube daily, and can accommodate up to 1000 attendees. Attendance has topped over 600 and averages 200 to 300 people daily; this has been sustained for 10 weeks. In addition, there is a weekly Thursday trainee series that hosts three 20‐minute seminars. In this forum, we reflect on a series that allowed global scientists to come together to help shape chaos into an opportunity for community and growth.
- Published
- 2020
- Full Text
- View/download PDF
6. Mature murine megakaryocytes present antigen-MHC class I molecules to T cells and transfer them to platelets
- Author
-
Anne Zufferey, Edwin R. Speck, Kellie R. Machlus, Rukhsana Aslam, Li Guo, Mark J. McVey, Michael Kim, Rick Kapur, Eric Boilard, Joseph E. Italiano, Jr, and John W. Semple
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Megakaryocytes (MKs) are bone marrow–derived cells that are primarily responsible for generating platelets for the maintenance of hemostasis. Although MK can variably express major histocompatibility complex (MHC) class I and II molecules during their differentiation, little is known whether they can elicit nonhemostatic immune functions such as T-cell activation. Here, we demonstrate that mature CD34− MHC class II− CD41+ MKs can endocytose exogenous ovalbumin (OVA) and proteolytically generate its immunogenic peptide ligand, which is crosspresented on their surface in association with MHC class I molecules. This crosspresentation triggered in vitro and in vivo OVA-specific CD8+ T-cell activation and proliferation. In addition, the OVA-MHC class I complexes were transferred from MK to pro-platelets upon thrombopoiesis in vitro. MK could also present endogenous MK-associated (CD61) peptides to activate CD61-specific CD8+ T cells and mediate immune thrombocytopenia in vivo. These results suggest that, in addition to their hemostatic role, mature MKs can significantly affect antigen-specific CD8+ T-cell responses via antigen presentation and are able to spread this immunogenic information through platelets.
- Published
- 2017
- Full Text
- View/download PDF
7. Deletion of the Arp2/3 complex in megakaryocytes leads to microthrombocytopenia in mice
- Author
-
David S. Paul, Caterina Casari, Congying Wu, Raymond Piatt, Swetha Pasala, Robert A. Campbell, Kathryn O. Poe, Dorsaf Ghalloussi, Robert H. Lee, Jeremy D. Rotty, Brian C. Cooley, Kellie R. Machlus, Joseph E. Italiano, Jr, Andrew S. Weyrich, James E. Bear, and Wolfgang Bergmeier
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Actin reorganization regulates key processes in platelet activation. Here we examined the role of the Arp2/3 complex, an essential component in actin filament branching, in platelet function. The Arpc2 gene, encoding the p34 subunit of the Arp2/3 complex, was deleted in the megakaryocyte lineage (Arpc2fl/flPF4-Cre). Deletion of the Arp2/3 complex resulted in marked microthrombocytopenia in mice, caused by premature platelet release into the bone marrow compartment and impaired platelet survival in circulation. Arpc2fl/flPF4-Cre platelets exhibited alterations in their actin cytoskeleton and their peripheral microtubule coil. Thrombocytopenia was alleviated following clodronate liposome-induced macrophage depletion in Arpc2fl/flPF4-Cre mice. Arpc2fl/flPF4-Cre platelets failed to spread and showed a mild defect in integrin activation and aggregation; however, no significant differences in hemostasis or thrombosis were observed between Arpc2fl/flPF4-Cre and control mice. Thus, Arp2/3 is critical for platelet homeostasis but plays only a minor role for vascular hemostasis.
- Published
- 2017
- Full Text
- View/download PDF
8. Scalable Generation of Universal Platelets from Human Induced Pluripotent Stem Cells
- Author
-
Qiang Feng, Namrata Shabrani, Jonathan N. Thon, Hongguang Huo, Austin Thiel, Kellie R. Machlus, Kyungho Kim, Julie Brooks, Feng Li, Chenmei Luo, Erin A. Kimbrel, Jiwu Wang, Kwang-Soo Kim, Joseph Italiano, Jaehyung Cho, Shi-Jiang Lu, and Robert Lanza
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Human induced pluripotent stem cells (iPSCs) provide a potentially replenishable source for the production of transfusable platelets. Here, we describe a method to generate megakaryocytes (MKs) and functional platelets from iPSCs in a scalable manner under serum/feeder-free conditions. The method also permits the cryopreservation of MK progenitors, enabling a rapid “surge” capacity when large numbers of platelets are needed. Ultrastructural/morphological analyses show no major differences between iPSC platelets and human blood platelets. iPSC platelets form aggregates, lamellipodia, and filopodia after activation and circulate in macrophage-depleted animals and incorporate into developing mouse thrombi in a manner identical to human platelets. By knocking out the β2-microglobulin gene, we have generated platelets that are negative for the major histocompatibility antigens. The scalable generation of HLA-ABC-negative platelets from a renewable cell source represents an important step toward generating universal platelets for transfusion as well as a potential strategy for the management of platelet refractoriness.
- Published
- 2014
- Full Text
- View/download PDF
9. Supplementary Information from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Materials and Methods, Supplementary Figures S1-S12
- Published
- 2023
- Full Text
- View/download PDF
10. Supp Table 3 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 3 HD and MPN samples.
- Published
- 2023
- Full Text
- View/download PDF
11. Supp Table 1 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 1 VEGFAC Top Differentially Expressed Genes by Cluster
- Published
- 2023
- Full Text
- View/download PDF
12. Supp Table 4 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
MM ALL Donor Details
- Published
- 2023
- Full Text
- View/download PDF
13. Supp Table 2 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 2 Gene sets for GSEA
- Published
- 2023
- Full Text
- View/download PDF
14. Supp Table 6 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 6 qRT PCR Probes
- Published
- 2023
- Full Text
- View/download PDF
15. Supp Table 5 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 5 Antibodies
- Published
- 2023
- Full Text
- View/download PDF
16. Supp Table 7 from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
Supplementary Table 7 NGS Panel
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Human Bone Marrow Organoids for Disease Modeling, Discovery, and Validation of Therapeutic Targets in Hematologic Malignancies
- Author
-
Bethan Psaila, Kellie R. Machlus, Julie Rayes, Sarah Gooding, Anindita Roy, Gowsihan Poologasundarampillai, David Bassett, Adam P. Croft, Adele K. Fielding, Emily A. Cutler, Samuel Kemble, Andrew P. Stone, Christina Simoglou Karali, Natalina E. Elliott, Rebecca E. Ling, Christopher B. Mahony, Gina Perrella, Beata Grygielska, Lauren C. Murphy, Nikolaos Sousos, Wei Xiong Wen, Guanlin Wang, Michela Colombo, Aude-Anais Olijnik, Jasmeet S. Reyat, Antonio Rodriguez-Romera, and Abdullah O. Khan
- Abstract
A lack of models that recapitulate the complexity of human bone marrow has hampered mechanistic studies of normal and malignant hematopoiesis and the validation of novel therapies. Here, we describe a step-wise, directed-differentiation protocol in which organoids are generated from induced pluripotent stem cells committed to mesenchymal, endothelial, and hematopoietic lineages. These 3D structures capture key features of human bone marrow—stroma, lumen-forming sinusoids, and myeloid cells including proplatelet-forming megakaryocytes. The organoids supported the engraftment and survival of cells from patients with blood malignancies, including cancer types notoriously difficult to maintain ex vivo. Fibrosis of the organoid occurred following TGFβ stimulation and engraftment with myelofibrosis but not healthy donor–derived cells, validating this platform as a powerful tool for studies of malignant cells and their interactions within a human bone marrow–like milieu. This enabling technology is likely to accelerate the discovery and prioritization of novel targets for bone marrow disorders and blood cancers.Significance:We present a human bone marrow organoid that supports the growth of primary cells from patients with myeloid and lymphoid blood cancers. This model allows for mechanistic studies of blood cancers in the context of their microenvironment and provides a much-needed ex vivo tool for the prioritization of new therapeutics.See related commentary by Derecka and Crispino, p. 263.This article is highlighted in the In This Issue feature, p. 247
- Published
- 2023
- Full Text
- View/download PDF
18. Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36
- Author
-
Maria N Barrachina, Gerard Pernes, Isabelle C Becker, Isabelle Allaeys, Thomas I. Hirsch, Dafna J Groeneveld, Abdullah O. Khan, Daniela Freire, Karen Guo, Estelle Carminita, Pooranee K Morgan, Thomas J Collins, Natalie A Mellett, Zimu Wei, Ibrahim Almazni, Joseph E. Italiano, James Luyendyk, Peter J Meikle, Mark Puder, Neil V. Morgan, Eric Boilard, Andrew J Murphy, and Kellie R Machlus
- Subjects
Article - Abstract
Lipids contribute to hematopoiesis and membrane properties and dynamics, however, little is known about the role of lipids in megakaryopoiesis. Here, a lipidomic analysis of megakaryocyte progenitors, megakaryocytes, and platelets revealed a unique lipidome progressively enriched in polyunsaturated fatty acid (PUFA)-containing phospholipids. In vitro, inhibition of both exogenous fatty acid functionalization and uptake and de novo lipogenesis impaired megakaryocyte differentiation and proplatelet production. In vivo, mice on a high saturated fatty acid diet had significantly lower platelet counts, which was prevented by eating a PUFA-enriched diet. Fatty acid uptake was largely dependent on CD36, and its deletion in mice resulted in thrombocytopenia. Moreover, patients with a CD36 loss-of-function mutation exhibited thrombocytopenia and increased bleeding. Our results suggest that fatty acid uptake and regulation is essential for megakaryocyte maturation and platelet production, and that changes in dietary fatty acids may be a novel and viable target to modulate platelet counts.
- Published
- 2023
19. Spatial transcriptomics of murine bone marrow megakaryocytes at single-cell resolution
- Author
-
Julia Tilburg, Andrew P. Stone, James M. Billingsley, David K. Scoville, Anna Pavenko, Yan Liang, Joseph E. Italiano, and Kellie R. Machlus
- Subjects
Hematology - Published
- 2023
- Full Text
- View/download PDF
20. Fluorescence artifact correction in the thrombin generation assay: Necessity for correction algorithms in procoagulant samples
- Author
-
Mikhail V Ovanesov, William C. Chang, Joseph W Jackson, Alisa S. Wolberg, and Kellie R. Machlus
- Subjects
Chemistry ,lcsh:RC633-647.5 ,Substrate (chemistry) ,Hematology ,lcsh:Diseases of the blood and blood-forming organs ,Original Articles ‐ Thrombosis ,Blood coagulation factors ,Thrombomodulin ,blood coagulation factors ,calibration ,Thrombin generation ,Molecular biology ,Fluorescence ,thrombin ,Thrombin ,Coagulation ,Hemostasis ,medicine ,hemostasis ,Original Article ,plasma ,medicine.drug - Abstract
Introduction The thrombin generation (TG) test is a global hemostasis assay sensitive to procoagulant conditions. However, some TG assays may underestimate elevated TG when the thrombin fluorogenic substrate is depleted or fluorescence is attenuated by the inner filter effect (IFE). Objectives We sought to elucidate the extent to which procoagulant conditions require correcting for fluorogenic substrate depletion and/or IFE. Methods We analyzed corrections for substrate depletion and IFE and their effect on TG parameters in plasma samples with elevated blood coagulation factors in the presence or absence of thrombomodulin via commercial calibrated automated thrombogram (CAT) platform and in‐house software capable of internal thrombin calibration with or without CAT‐like artifact correction. Results Elevated thrombin peak height (TPH) and endogenous thrombin potential (ETP) were detected with 2× and 4× increases in blood coagulation factors I, V, VIII, IX, X, and XI, or prothrombin in the presence or absence of artifact correction. The effect of the CAT algorithm was evident in TG curves from both low procoagulant (thrombomodulin‐supplemented) and procoagulant (factor‐supplemented) plasma samples. However, in all samples, with the exception of elevated prothrombin, CAT’s correction was small (
- Published
- 2021
21. Beyond the thrombus: Platelet-inspired nanomedicine approaches in inflammation, immune response, and cancer
- Author
-
Cian Desai, Milka Koupenova, Kellie R. Machlus, and Anirban Sen Gupta
- Subjects
Blood Platelets ,Inflammation ,Nanomedicine ,Neoplasms ,Immunity ,Tumor Microenvironment ,Humans ,Thrombosis ,Hematology - Abstract
The traditional role of platelets is in the formation of blood clots for physiologic (e.g., in hemostasis) or pathologic (e.g., in thrombosis) functions. The cellular and subcellular mechanisms and signaling in platelets involved in these functions have been extensively elucidated and new knowledge continues to emerge, resulting in various therapeutic developments in this area for the management of hemorrhagic or thrombotic events. Nanomedicine, a field involving design of nanoparticles with unique biointeractive surface modifications and payload encapsulation for disease-targeted drug delivery, has become an important component of such therapeutic development. Beyond their traditional role in blood clotting, platelets have been implicated to play crucial mechanistic roles in other diseases including inflammation, immune response, and cancer, via direct cellular interactions, as well as secretion of soluble factors that aid in the disease microenvironment. To date, the development of nanomedicine systems that leverage these broader roles of platelets has been limited. Additionally, another exciting area of research that has emerged in recent years is that of platelet-derived extracellular vesicles (PEVs) that can directly and indirectly influence physiological and pathological processes. This makes PEVs a unique paradigm for platelet-inspired therapeutic design. This review aims to provide mechanistic insight into the involvement of platelets and PEVs beyond hemostasis and thrombosis, and to discuss the current state of the art in the development of platelet-inspired therapeutic technologies in these areas, with an emphasis on future opportunities.
- Published
- 2022
22. High‐content, label‐free analysis of proplatelet production from megakaryocytes
- Author
-
Shauna L. French, Adrian R. Wilkie, Benjamin Posorske, Prakrith Vijey, Anjana Ray, Lillian J. Horin, Kyle W. Karhohs, Joseph E. Italiano, Anne E. Carpenter, and Kellie R. Machlus
- Subjects
Blood Platelets ,Computer science ,Hematology ,Computational biology ,030204 cardiovascular system & hematology ,Thrombocytopenia ,Article ,Skeletonization ,Thrombopoiesis ,03 medical and health sciences ,Statistical classification ,0302 clinical medicine ,Open source ,Platelet production ,Humans ,Platelet formation ,Megakaryocytes ,Cells, Cultured ,Label free - Abstract
The mechanisms that regulate platelet biogenesis remain unclear; factors that trigger megakaryocytes (MKs) to initiate platelet production are poorly understood. Platelet formation begins with proplatelets which are cellular extensions that originate from the MK cell body. Proplatelet formation is a highly dynamic and asynchronous process which poses unique challenges for researchers to accurately capture and analyze. We have designed an open-source, high-content, high-throughput, label-free analysis platform. Phase-contrast images of live, primary MKs are captured over a 24-hour period. Pixel-based machine-learning classification done by ilastik generates probability maps of key cellular features (circular MKs and branching proplatelets), which are then processed by a customized CellProfiler pipeline to identify and filter structures of interest based on morphologic parameters. CellProfiler Analyst, provides a final, supervised, machine learning classification to bolster accurate identification of cellular structures. This entire workflow yields the percent of proplatelet production, area, and count of proplatelets and MKs, as well as other statistics including skeletonization information for measuring proplatelet branching and length. We propose using a combination of these analyzed metrics, in particular the area measurements of MKs and proplatelets, when assessing in-vitro proplatelet production. Accuracy was validated against manually counted images and an existing algorithm. We then used the new platform to test compounds known to cause thrombocytopenia, including bromodomain inhibitors, and uncovered previously unrecognized effects of drugs on proplatelet formation, thus demonstrating the utility of our analysis platform. This advance in creating unbiased data analysis will increase the scale and scope of proplatelet production studies and potentially serve as a valuable resource for investigating molecular mechanisms of thrombocytopenia.
- Published
- 2020
- Full Text
- View/download PDF
23. In vitro megakaryocyte culture from human bone marrow aspirates as a research and diagnostic tool
- Author
-
Elena Y Osipova, Kirill R Butov, Nikita B Mikhalkin, Mikhail A. Panteleev, Kellie R. Machlus, and Natalia M. Trubina
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,urogenital system ,Human bone ,Bone Marrow Cells ,Cell Differentiation ,Hematology ,General Medicine ,030204 cardiovascular system & hematology ,Biology ,Article ,In vitro ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Megakaryocyte ,Nucleated cell ,medicine ,Humans ,Platelet ,Bone marrow ,Megakaryocytes ,Cells, Cultured - Abstract
Megakaryocytes (MKs) are relatively rare in bone marrow, comprising
- Published
- 2020
- Full Text
- View/download PDF
24. Platelet-derived extracellular vesicles infiltrate and modify the bone marrow during inflammation
- Author
-
Patricia Davenport, Isabelle Allaeys, Jorge Canas, Shauna L. French, Audrée Laroche, Natalia M. Trubina, Marsha A. Moses, Mikhail A. Panteleev, Kellie R. Machlus, Kirill R Butov, Golnaz Morad, Eric Boilard, Martha Sola-Visner, and Joseph E. Italiano
- Subjects
Blood Platelets ,0301 basic medicine ,Inflammation ,030204 cardiovascular system & hematology ,Systemic inflammation ,Flow cytometry ,Extracellular Vesicles ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,medicine ,Animals ,Platelet ,Platelet activation ,medicine.diagnostic_test ,Chemistry ,fungi ,Hematology ,Platelets and Thrombopoiesis ,medicine.disease ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Bone marrow ,medicine.symptom ,Megakaryocytes ,Infiltration (medical) - Abstract
During inflammation, steady-state hematopoiesis switches to emergency hematopoiesis to repopulate myeloid cells, with a bias toward the megakaryocytic lineage. Soluble inflammatory cues are thought to be largely responsible for these alterations. However, how these plasma factors rapidly alter the bone marrow (BM) is not understood. Inflammation also drives platelet activation, causing the release of platelet-derived extracellular vesicles (PEVs), which package diverse cargo and reprogram target cells. We hypothesized that PEVs infiltrate the BM, providing a direct mode of communication between the plasma and BM environments. We transfused fluorescent, wild-type (MPL+) platelets into recipient cMpl−/−mice before triggering systemic inflammation. Twenty hours postinfusion, we observed significant infiltration of donor platelet-derived particles in the BM, which we tracked immunophenotypically (MPL+ immunohistochemistry staining) and quantified by flow cytometry. To determine if this phenomenon relates to humans, we extensively characterized both megakaryocyte-derived and PEVs generated in vitro and in vivo, and found enrichment of extracellular vesicles in bone marrow compared with autologous peripheral blood. Last, BM from cMpl−/− mice was cultured in the presence or absence of wild-type (MPL+) PEVs. After 72 hours, flow cytometry revealed increased megakaryocytes only in cultures with added PEVs. The majority of CD41+ cells were bound to PEVs, suggesting a PEV-mediated rescue of megakaryopoiesis. In conclusion, we report for the first time that plasma-residing PEVs infiltrate the BM. Further, PEVs interact with BM cells in vivo and in vitro, causing functional reprogramming that may represent a novel model of inflammation-induced hematopoiesis.
- Published
- 2020
- Full Text
- View/download PDF
25. Human bone marrow organoids for disease modelling, discovery and validation of therapeutic targets in hematological malignancies
- Author
-
Abdullah O. Khan, Michela Colombo, Jasmeet S. Reyat, Guanlin Wang, Antonio Rodriguez-Romera, Wei Xiong Wen, Lauren Murphy, Beata Grygielska, Chris Mahoney, Andrew Stone, Adam Croft, David Bassett, Gowsihan Poologasundarampillai, Anindita Roy, Sarah Gooding, Julie Rayes, Kellie R Machlus, and Bethan Psaila
- Abstract
A lack of models that recapitulate the complexity of human bone marrow has hampered mechanistic studies of normal and malignant hematopoiesis and the validation of novel therapies. Here, we describe a step-wise, directed-differentiation protocol in which organoids are generated from iPSCs committed to mesenchymal, endothelial and hematopoietic lineages. These 3-dimensional structures capture key features of human bone marrow - stroma, lumen-forming sinusoidal vessels and myeloid cells including pro-platelet forming megakaryocytes. The organoids supported the engraftment and survival of cells from patients with blood malignancies, including cancer types notoriously difficult to maintain ex vivo. Fibrosis of the organoid occurred following TGFβ stimulation and engraftment with myelofibrosis but not healthy donor-derived cells, validating this platform as a powerful tool for studies of malignant cells and their interactions within a human bone marrow-like milieu. This enabling technology is likely to accelerate discovery and prioritization of novel targets for bone marrow disorders and blood cancers.Significance StatementWe present a 3D, vascularised human bone marrow organoid that supports growth of primary cells from patients with myeloid and lymphoid blood cancers. This model allows for mechanistic studies of blood cancers in the context of their microenvironment, and provides a much-needed, ex vivo tool for prioritization of new therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
26. The origin of the megakaryocyte
- Author
-
Kellie R. Machlus and Eric Boilard
- Published
- 2022
- Full Text
- View/download PDF
27. Thrombocytopenia in a Mouse Model of Pancreatic Cancer
- Author
-
Michelle M. Luo, Yaqiu Sang, Maria N. Barrachina, Andrew P. Stone, Keely G. Davey, Yi Yang, Lori A. Holle, Tomohiro Kawano, Nigel Mackman, Matthew J. Flick, Kellie R. Machlus, and Alisa S. Wolberg
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
28. Illustrated State-of-the-Art Capsules of the ISTH 2021 Congress
- Author
-
Marguerite Neerman-Arbez, Marc Carrier, Kellie R. Machlus, Laurence Panicot-Dubois, Coen Maas, Andra H. James, Weikai Li, Colin A. Kretz, Kathleen Freson, Tiziano Barbui, Audrey C. A. Cleuren, Jamie M. O’Sullivan, Paul Clinton Spiegel, Frank W.G. Leebeek, Michael Makris, Laura E. Green, Philip J. Hogg, Jane E. Freedman, Suzanne C. Cannegieter, Anetta Undas, Michelle Lavin, Simon J. Stanworth, James S. O’Donnell, Peter William Collins, Yaseen M. Arabi, Sriram Krishnaswamy, Verena Schroeder, Madhvi Rajpurkar, Leonid Medved, Walter Ageno, and Ida Martinelli
- Subjects
education.field_of_study ,Medical education ,Coronavirus disease 2019 (COVID-19) ,business.industry ,media_common.quotation_subject ,Platelet disorder ,Population ,education ,Vascular biology ,Hematology ,State (polity) ,SDG 3 - Good Health and Well-being ,Educational resources ,Medicine ,Diseases of the blood and blood-forming organs ,Social media ,RC633-647.5 ,business ,Venous thromboembolism ,media_common - Abstract
This year's Congress of the International Society of Thrombosis and Haemostasis (ISTH) was hosted virtually from Philadelphia July 17-21, 2021. The conference, now held annually, highlighted cutting-edge advances in basic, population and clinical sciences of relevance to the Society. Despite being held virtually, the 2021 congress was of the same scope and quality as an annual meeting held in person. An added feature of the program is that talks streamed at the designated times will then be available on-line for asynchronous viewing. The program included 77 State of the Art (SOA) talks, thematically grouped in 28 sessions, given by internationally recognized leaders in the field. The SOA speakers were invited to prepare brief illustrated reviews of their talks that were peer reviewed and are included in this article. The topics, across the main scientific themes of the congress, include Arterial Thromboembolism, Coagulation and Natural Anticoagulants, COVID-19 and Coagulation, Diagnostics and Omics, Fibrinogen, Fibrinolysis and Proteolysis, Hemophilia and Rare Bleeding Disorders, Hemostasis in Cancer, Inflammation and Immunity, Pediatrics, Platelet Disorders, von Willebrand Disease and Thrombotic Angiopathies, Platelets and Megakaryocytes, Vascular Biology, Venous Thromboembolism and Women's Health. These illustrated capsules highlight the major scientific advances with potential to impact clinical practice. Readers are invited to take advantage of the excellent educational resource provided by these illustrated capsules. They are also encouraged to use the image in social media to draw attention to the high quality and impact of the science presented at the congress. ispartof: RESEARCH AND PRACTICE IN THROMBOSIS AND HAEMOSTASIS vol:5 issue:5 ispartof: location:United States status: published
- Published
- 2021
- Full Text
- View/download PDF
29. Modulation of megakaryopoiesis and platelet production during inflammation
- Author
-
Kellie R. Machlus and Genevieve Couldwell
- Subjects
Blood Platelets ,Inflammation ,urogenital system ,Hematopoietic stem cell ,Cell Differentiation ,Hematology ,030204 cardiovascular system & hematology ,Biology ,Hematopoiesis ,Proinflammatory cytokine ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,medicine.anatomical_structure ,Megakaryocyte ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Humans ,Platelet ,Bone marrow ,medicine.symptom ,Progenitor cell ,Megakaryocytes - Abstract
Megakaryocytes (MKs) are widely known as the progenitor cells of platelets. These large, polyploid cells are a derivative of the hematopoietic stem cell (HSC), and reside in the bone marrow, lining blood vessel walls where they release their platelet progeny into circulation. Although little is known about how MKs differ under various environmental stressors, both chronic and acute inflammation alter the differentiation and molecular content of MKs. Furthermore, evidence suggests that the release of inflammatory cytokines may induce MK rupture and rapid release of platelets as a mechanism to quickly replenish diminished platelet counts in response to inflammation. Similarities between MKs and their close relatives, white blood cells, have introduced the notion that MKs may play a role in combating infection by engulfing and presenting antigens, and passing this information to circulating platelets. In addition, MKs exposed to varying bone marrow environments produce different platelets which enter circulation primed to respond to and combat inflammation, infection, or injury. This review focuses on how inflammation alters MK production, maturation, and platelet production. In addition, it introduces the idea that inflammation reprograms MKs to create different, more pathogenic platelets and leads them to take on different roles as responders to deleterious conditions. In the future, studies determining how platelets are altered in disease states may lead to novel MK- and platelet-based therapeutic targets to mitigate inflammation-related morbidity and mortality.
- Published
- 2019
- Full Text
- View/download PDF
30. Location is everything when it comes to megakaryocyte function
- Author
-
Kellie R. Machlus and Eric Boilard
- Subjects
Blood Platelets ,0301 basic medicine ,Population ,Biology ,Thrombopoiesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Megakaryocyte ,Antigen ,Bone Marrow ,medicine ,Animals ,Platelet ,education ,education.field_of_study ,urogenital system ,Cell Differentiation ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Phenotype ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,bacteria ,Bone marrow ,Megakaryocytes ,Function (biology) ,Research Article - Abstract
Although platelets are the cellular mediators of thrombosis, they are also immune cells. Platelets interact both directly and indirectly with immune cells, impacting their activation and differentiation, as well as all phases of the immune response. Megakaryocytes (Mks) are the cell source of circulating platelets, and until recently Mks were typically only considered bone marrow–resident (BM-resident) cells. However, platelet-producing Mks also reside in the lung, and lung Mks express greater levels of immune molecules compared with BM Mks. We therefore sought to define the immune functions of lung Mks. Using single-cell RNA sequencing of BM and lung myeloid-enriched cells, we found that lung Mks, which we term Mk(L), had gene expression patterns that are similar to antigen-presenting cells. This was confirmed using imaging and conventional flow cytometry. The immune phenotype of Mks was plastic and driven by the tissue immune environment, as evidenced by BM Mks having an Mk(L)-like phenotype under the influence of pathogen receptor challenge and lung-associated immune molecules, such as IL-33. Our in vitro and in vivo assays demonstrated that Mk(L) internalized and processed both antigenic proteins and bacterial pathogens. Furthermore, Mk(L) induced CD4(+) T cell activation in an MHC II–dependent manner both in vitro and in vivo. These data indicated that Mk(L) had key immune regulatory roles dictated in part by the tissue environment.
- Published
- 2021
- Full Text
- View/download PDF
31. Platelet Extracellular Vesicles: Beyond the Blood
- Author
-
Eric Boilard, Florian Puhm, and Kellie R. Machlus
- Subjects
0301 basic medicine ,Blood Platelets ,Inflammation ,Cell Communication ,030204 cardiovascular system & hematology ,Extracellular vesicles ,Article ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Cell-Derived Microparticles ,Organelle ,Synovial Fluid ,medicine ,Synovial fluid ,Animals ,Humans ,Platelet ,Hemostasis ,Chemistry ,Platelet Activation ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Coagulation ,Bone marrow ,Lymph ,medicine.symptom ,Inflammation Mediators ,Cardiology and Cardiovascular Medicine - Abstract
Extracellular vesicles (EVs) are a means of cell-to-cell communication and can facilitate the exchange of a broad array of molecules between adjacent or distant cells. Platelets are anucleate cells derived from megakaryocytes and are primarily known for their role in maintaining hemostasis and vascular integrity. Upon activation by a variety of agonists, platelets readily generate EVs, which were initially identified as procoagulant particles. However, as both platelets and their EVs are abundant in blood, the role of platelet EVs in hemostasis may be redundant. Moreover, findings have challenged the significance of platelet-derived EVs in coagulation. Looking beyond hemostasis, platelet EV cargo is incredibly diverse and can include lipids, proteins, nucleic acids, and organelles involved in numerous other biological processes. Furthermore, while platelets cannot cross tissue barriers, their EVs can enter lymph, bone marrow, and synovial fluid. This allows for the transfer of platelet-derived content to cellular recipients and organs inaccessible to platelets. This review highlights the importance of platelet-derived EVs in physiological and pathological conditions beyond hemostasis.
- Published
- 2020
32. Comparative Analysis of Thrombin Calibration Algorithms and Correction for Thrombin-α2macroglobulin Activity
- Author
-
Joseph W Jackson, Alisa S. Wolberg, Mikhail V Ovanesov, William C. Chang, and Kellie R. Machlus
- Subjects
Normalization (statistics) ,genetic structures ,lcsh:Medicine ,030204 cardiovascular system & hematology ,Thrombomodulin ,Article ,03 medical and health sciences ,Tissue factor ,0302 clinical medicine ,Thrombin ,medicine ,Calibration ,plasma ,Blood coagulation test ,business.industry ,lcsh:R ,Significant difference ,blood coagulation tests ,General Medicine ,blood coagulation factors ,calibration ,thrombin ,030220 oncology & carcinogenesis ,Filter effect ,business ,Algorithm ,medicine.drug ,circulatory and respiratory physiology - Abstract
Background: The thrombin generation (TG) test is useful for characterizing global hemostasis potential, but fluorescence substrate artifacts, such as thrombin-&alpha, 2macroglobulin (T-&alpha, 2MG) signal, inner filter effect (IFE), substrate consumption, and calibration algorithms have been suggested as sources of intra- and inter-laboratory variance, which may limit its clinical utility. Methods: Effects of internal vs. external normalization, IFE and T-&alpha, 2MG on TG curves in normal plasma supplemented with coagulation factors, thrombomodulin, and tissue factor were studied using the Calibrated Automated Thrombinography (CAT, Diagnostica Stago, Parsippany, NJ, USA) and in-house software. Results: The various calibration methods demonstrated no significant difference in producing TG curves, nor increased the robustness of the TG assay. Several TG parameters, including thrombin peak height (TPH), produced from internal linear calibration did not differ significantly from uncalibrated TG parameters. Further, TPH values from internal linear and nonlinear calibration with or without T-&alpha, 2MG correction correlated well with TPH from external calibration. Higher coefficients of variation (CVs) for TPH values were observed in both platelet-free and platelet-rich plasma with added thrombomodulin. Conclusions: Our work suggests minimal differences between distinct computational approaches toward calibrating and correcting fluorescence signals into TG levels, with most samples returning similar or equivalent TPH results.
- Published
- 2020
- Full Text
- View/download PDF
33. Novel gene variants in patients with platelet-based bleeding using combined exome sequencing and RNAseq murine expression data
- Author
-
Jasmeet S. Reyat, Rachel J Stapley, Neil V. Morgan, Susanne N. Wijesinghe, Ibrahim Abdullah F Almazni, Kellie R. Machlus, Jeremy A. Pike, and Abdullah O. Khan
- Subjects
Blood Platelets ,Candidate gene ,Platelet disorder ,Functional testing ,Hemorrhage ,Hematology ,Computational biology ,030204 cardiovascular system & hematology ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cohort ,Mutation ,Exome Sequencing ,Animals ,Humans ,Platelet ,Exome ,Gene ,Genotyping ,Exome sequencing - Abstract
Essentials Identifying genetic variants in platelet disorders is challenging due to its heterogenous nature. We combine WES, RNAseq, and python-based bioinformatics to identify novel gene variants. We find novel candidates in patient data by cross-referencing against a murine RNAseq model of thrombopoiesis. This innovative combined bioinformatic approach provides novel data for future research in the field. ABSTRACT: Background The UK Genotyping and Phenotyping of Platelets study has recruited and analyzed 129 patients with suspected heritable bleeding. Previously, 55 individuals had a definitive genetic diagnosis based on whole exome sequencing (WES) and platelet morphological and functional testing. A significant challenge in this field is defining filtering criteria to identify the most likely candidate mutations for diagnosis and further study. Objective Identify candidate gene mutations for the remaining 74 patients with platelet-based bleeding with unknown genetic cause, forming the basis of future re-recruitment and further functional testing and assessment. Methods Using python-based data frame indexing, we first identify and filter all novel and rare variants using a panel of 116 genes known to cause bleeding across the full cohort of WES data. This identified new variants not previously reported in this cohort. We then index the remaining patients, with rare or novel variants in known bleeding genes against a murine RNA sequencing dataset that models proplatelet-forming megakaryocytes. Results Filtering against known genes identified candidate variants in 59 individuals, including novel variants in several known genes. In the remaining cohort of "unknown" patients, indexing against differentially expressed genes revealed candidate gene variants in several novel unreported genes, focusing on 14 patients with a severe clinical presentation. Conclusions We identified candidate mutations in a cohort of patients with no previous genetic diagnosis. This work involves innovative coupling of RNA sequencing and WES to identify candidate variants forming the basis of future study in a significant number of undiagnosed patients.
- Published
- 2020
34. FcγRIIA expression accelerates nephritis and increases platelet activation in systemic lupus erythematosus
- Author
-
Robert A. Campbell, Joyce Rauch, Benoit Mailhot, Paul R. Fortin, Eric Boilard, David Salem, Steve Lacroix, Christian Lood, Audrée Laroche, Matthew T. Rondina, Isabelle Allaeys, Nathalie Cloutier, Nicolas Tessandier, Kellie R. Machlus, Tania Lévesque, Steven E. McKenzie, Anne Zufferey, Imene Melki, Natalie Patey, Jesse W. Rowley, and Arnaud Droit
- Subjects
Blood Platelets ,Immunology ,Lupus nephritis ,Mice, Transgenic ,Biochemistry ,Immunoglobulin G ,Mice ,Immune system ,immune system diseases ,medicine ,Animals ,Platelet ,Platelet activation ,skin and connective tissue diseases ,Autoantibodies ,Systemic lupus erythematosus ,biology ,business.industry ,Receptors, IgG ,Cell Biology ,Hematology ,medicine.disease ,Platelet Activation ,Platelets and Thrombopoiesis ,Lupus Nephritis ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,Bone marrow ,business ,Nephritis - Abstract
Systemic lupus erythematosus (SLE) is an autoimmune inflammatory disease characterized by deposits of immune complexes (ICs) in organs and tissues. The expression of FcγRIIA by human platelets, which is their unique receptor for immunoglobulin G antibodies, positions them to ideally respond to circulating ICs. Whereas chronic platelet activation and thrombosis are well-recognized features of human SLE, the exact mechanisms underlying platelet activation in SLE remain unknown. Here, we evaluated the involvement of FcγRIIA in the course of SLE and platelet activation. In patients with SLE, levels of ICs are associated with platelet activation. Because FcγRIIA is absent in mice, and murine platelets do not respond to ICs in any existing mouse model of SLE, we introduced the FcγRIIA (FCGR2A) transgene into the NZB/NZWF1 mouse model of SLE. In mice, FcγRIIA expression by bone marrow cells severely aggravated lupus nephritis and accelerated death. Lupus onset initiated major changes to the platelet transcriptome, both in FcγRIIA-expressing and nonexpressing mice, but enrichment for type I interferon response gene changes was specifically observed in the FcγRIIA mice. Moreover, circulating platelets were degranulated and were found to interact with neutrophils in FcγRIIA-expressing lupus mice. FcγRIIA expression in lupus mice also led to thrombosis in lungs and kidneys. The model recapitulates hallmarks of human SLE and can be used to identify contributions of different cellular lineages in the manifestations of SLE. The study further reveals a role for FcγRIIA in nephritis and in platelet activation in SLE.
- Published
- 2020
35. Human platelets express endothelial protein C receptor, which can be utilized to enhance localization of factor VIIa activity
- Author
-
Mirella Ezban, Kellie R. Machlus, Ammon M. Fager, and Maureane Hoffman
- Subjects
0301 basic medicine ,Endothelial protein C receptor ,Immunoprecipitation ,Chemistry ,Hematology ,030204 cardiovascular system & hematology ,Cell biology ,03 medical and health sciences ,Tissue factor ,030104 developmental biology ,0302 clinical medicine ,Coagulation ,Hemostasis ,medicine ,Platelet ,Platelet activation ,Protein C ,medicine.drug - Abstract
Essentials Factor VIIa binds activated platelets to promote hemostasis in hemophilia patients with inhibitors. The interactions and sites responsible for platelet-FVIIa binding are not fully understood. Endothelial cell protein C receptor (EPCR) is expressed on activated human platelets. EPCR binding enhances the efficacy of a FVIIa variant and could impact design of new therapeutics. Summary Background High-dose factor VIIa (FVIIa) is routinely used as an effective bypassing agent to treat hemophilia patients with inhibitory antibodies that compromise factor replacement. However, the mechanism by which FVIIa binds activated platelets to promote hemostasis is not fully understood. FVIIa-DVQ is an analog of FVIIa with enhanced tissue factor (TF)-independent activity and hemostatic efficacy relative to FVIIa. Our previous studies have shown that FVIIa-DVQ exhibits greater platelet binding, thereby suggesting that features in addition to lipid composition contribute to platelet-FVIIa interactions. Objectives Endothelial cell protein C receptor (EPCR) also functions as a receptor for FVIIa on endothelial cells. We therefore hypothesized that an interaction with EPCR might play a role in platelet-FVIIa binding. Methods/results In the present study, we used flow cytometric analyses to show that platelet binding of both FVIIa and FVIIa-DVQ is partially inhibited in the presence of excess protein C or an anti-EPCR antibody. This decreased binding results in a corresponding decrease in the activity of both molecules in FXa and thrombin generation assays. Enhanced binding to EPCR was sufficient to account for the increased platelet binding of FVIIa-DVQ compared with wild-type FVIIa. As EPCR protein expression has not previously been shown in platelets, we confirmed the presence of EPCR in platelets using immunofluorescence, flow cytometry, immunoprecipitation, and mass spectrometry. Conclusions This work represents the first demonstration that human platelets express EPCR and suggests that modulation of EPCR binding could be utilized to enhance the hemostatic efficacy of rationally designed FVIIa analogs.
- Published
- 2018
- Full Text
- View/download PDF
36. Tyrosyl‐tRNA synthetase drives megakaryopoiesis independently of thrombopoietin signaling
- Author
-
Shauna L. French and Kellie R. Machlus
- Subjects
tyrosyl-tRNA synthetase ,Extramural ,Chemistry ,thrombocytopenia ,Cell Biology ,Hematology ,Biological Sciences ,Thrombopoiesis ,Cell biology ,Tyrosine—tRNA ligase ,Thrombopoietin ,PNAS Plus ,Tyrosine-tRNA Ligase ,Tyrosyl-tRNA Synthetase ,Signal transduction ,megakaryopoiesis ,Signal Transduction ,Megakaryopoiesis - Abstract
Significance Aminoacyl-tRNA synthetases (aaRSs) catalyze aminoacylation of tRNAs in the first step of protein synthesis in the cytoplasm. However, in higher eukaryotes, they acquired additional functions beyond translation. In the present study, we show that an activated form of tyrosyl-tRNA synthetase (YRSACT) functions to enhance megakaryopoiesis and platelet production in vitro and in vivo. These findings were confirmed with human megakaryocytes differentiated from peripheral blood CD34+ hematopoietic stem cells and with human induced pluripotent stem (iPS) cells. The activity of YRSACT is independent of thrombopoietin (TPO), as evidenced by expansion of the megakaryocytes from iPS cell-derived hematopoietic stem cells from a patient deficient in TPO signaling. These findings demonstrate a previously unrecognized function of an aaRS which may have implications for therapeutic interventions., New mechanisms behind blood cell formation continue to be uncovered, with therapeutic approaches for hematological diseases being of great interest. Here we report an enzyme in protein synthesis, known for cell-based activities beyond translation, is a factor inducing megakaryocyte-biased hematopoiesis, most likely under stress conditions. We show an activated form of tyrosyl-tRNA synthetase (YRSACT), prepared either by rationally designed mutagenesis or alternative splicing, induces expansion of a previously unrecognized high-ploidy Sca-1+ megakaryocyte population capable of accelerating platelet replenishment after depletion. Moreover, YRSACT targets monocytic cells to induce secretion of transacting cytokines that enhance megakaryocyte expansion stimulating the Toll-like receptor/MyD88 pathway. Platelet replenishment by YRSACT is independent of thrombopoietin (TPO), as evidenced by expansion of the megakaryocytes from induced pluripotent stem cell-derived hematopoietic stem cells from a patient deficient in TPO signaling. We suggest megakaryocyte-biased hematopoiesis induced by YRSACT offers new approaches for treating thrombocytopenia, boosting yields from cell-culture production of platelet concentrates for transfusion, and bridging therapy for hematopoietic stem cell transplantation.
- Published
- 2019
- Full Text
- View/download PDF
37. Automatic segmentation and analysis of fibrin networks in 3D confocal microscopy images.
- Author
-
Xiaomin Liu, Jian Mu, Kellie R. Machlus, Alisa S. Wolberg, Elliot D. Rosen, Zhiliang Xu, Mark S. Alber, and Danny Z. Chen
- Published
- 2012
- Full Text
- View/download PDF
38. Manipulation of Fatty Acid Metabolism Impairs Megakaryocyte Differentiation and Platelet Production
- Author
-
Andrew J. Murphy, Graeme I. Lancaster, Dafna J. Groeneveld, Isabelle C Becker, Gerard Pernes, Kellie R. Machlus, Maria N. Barrachina, Peter J. Meikle, and James P. Luyendyk
- Subjects
chemistry.chemical_compound ,Fatty acid metabolism ,Chemistry ,Megakaryocyte differentiation ,Immunology ,Platelet production ,Cell Biology ,Hematology ,Biochemistry ,Cell biology - Abstract
Background: Obesity is associated with an altered plasma lipid composition that impacts platelet activation, indicating that lipids directly affect platelet function and reactivity. However, very little is known about which essential lipids are important to enable platelet production from their precursors, megakaryocytes (MKs), and whether obesity influences lipid composition and consequently affects thrombopoiesis. Aim: We aimed to determine which lipids and associated pathways play a significant role in MK differentiation and platelet production in health and upon obesity. Our long-term goal is to determine if we can manipulate MK maturation and platelet production through lipid incorporation or inhibition. Methods: In order to determine the lipid profile of MKs through maturation, we performed an extensive lipidomics screen on primary MK progenitors, mature MKs, and platelets. Based on our findings, we then targeted de novo fatty acid (FA) synthesis in MKs to determine whether manipulation of this pathway attenuates MK differentiation and platelet formation. In parallel, we investigated megakaryo- and thrombopoiesis in an experimental setting of diet-induced obesity (DIO) in mice to examinate how altered lipid compositions within the plasma affect MK maturation. Results: Lipidomic data revealed increased polyunsaturated FA and plasmalogen content in the late stages of MKs maturation, suggesting that lipid composition is undergoing changes that may enable platelet production. To validate and extend these results, we inhibited multiple enzymes in de novo lipogenesis and FA synthesis. Using two different sources of primary murine hematopoietic stem cells (HSCs), we found that MK differentiation and maturation from HSCs was significantly decreased up to 60% after inhibition of acetyl-coA carboxylase (ACC) and 80% after fatty acid synthetase (FASN) inhibition, both enzymes from the de novo lipogenesis pathway. Moreover, we also found that MK differentiation and maturation were highly decreased (up to 70%) after the inhibition of acyl-coA synthetase (ACS). To further examine the role of fatty acids incorporation on platelet production, we next treated mature MKs with an ACS inhibitor to investigate the direct effect of FA incorporation on proplatelet formation; we observed a significant, multifoldreduction in proplatelet area. While dihydroceramides were altered in the lipidomic data, impairment of the de novo ceramide synthesis pathway did not affect either MK maturation or platelet production. Moreover, we observed an increased abundance of HSCs, myeloid cells, and MK progenitors in the bone marrow of DIO mice compared to control mice on a chow diet. This further supports an important role for lipids in megakaryopoiesis and suggests that differences in platelet reactivity during obesity may be caused by altered MK maturation and/or platelet production. Conclusions: Our results elucidate an important and previously unrecognized role for fatty acid synthesis in megakaryopoiesis and platelet production. The findings imply that an altered lipid content, as observed in patients with obesity, may not only impact platelet reactivity but also MK maturation and function. Consequently, platelets with an altered lipid content might originate from pathogenic MKs in obesity, which makes lipid-regulating proteins in MKs novel and viable therapeutic targets. Disclosures Machlus: KEROS Therapeutics: Consultancy, Honoraria; STRM.BIO: Consultancy, Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
39. New Insights into the Differentiation of Megakaryocytes from Hematopoietic Progenitors
- Author
-
Kellie R. Machlus, Shauna L. French, and Leila Noetzli
- Subjects
0301 basic medicine ,Cell signaling ,Context (language use) ,Bone Marrow Cells ,Cell Communication ,Biology ,Article ,Thrombopoiesis ,03 medical and health sciences ,0302 clinical medicine ,Megakaryocyte ,medicine ,Humans ,Cell Lineage ,Progenitor cell ,Megakaryopoiesis ,Inflammation ,Cell Cycle ,Hematopoietic stem cell ,Cell Differentiation ,Hematopoietic Stem Cells ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Thrombopoietin ,030220 oncology & carcinogenesis ,Stem cell ,Cardiology and Cardiovascular Medicine ,Megakaryocytes ,Signal Transduction ,Transcription Factors - Abstract
Megakaryocytes are hematopoietic cells, which are responsible for the production of blood platelets. The traditional view of megakaryopoiesis describes the cellular journey from hematopoietic stem cells, through a hierarchical series of progenitor cells, ultimately to a mature megakaryocyte. Once mature, the megakaryocyte then undergoes a terminal maturation process involving multiple rounds of endomitosis and cytoplasmic restructuring to allow platelet formation. However, recent studies have begun to redefine this hierarchy and shed new light on alternative routes by which hematopoietic stem cells are differentiated into megakaryocytes. In particular, the origin of megakaryocytes, including the existence and hierarchy of megakaryocyte progenitors, has been redefined, as new studies are suggesting that hematopoietic stem cells originate as megakaryocyte-primed and can bypass traditional lineage checkpoints. Overall, it is becoming evident that megakaryopoiesis does not only occur as a stepwise process, but is dynamic and adaptive to biological needs. In this review, we will reexamine the canonical dogmas of megakaryopoiesis and provide an updated framework for interpreting the roles of traditional pathways in the context of new megakaryocyte biology. Visual Overview— An online visual overview is available for this article.
- Published
- 2019
40. Author response: Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets
- Author
-
Rim Bouslama, Pierre Cunin, Andrew S. Weyrich, Allyn Morris, Eric Boilard, Pui Y. Lee, Marta Martínez-Bonet, Nathan Nelson-Maney, Martha Sola-Visner, Peter A. Nigrovic, Kellie R. Machlus, Li Guo, Alexandra Wactor, and Joseph E. Italiano
- Subjects
Emperipolesis ,Membrane ,medicine.anatomical_structure ,Megakaryocyte ,Chemistry ,medicine ,Platelet ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
41. Contributors
- Author
-
Joseph Alsousou, Dominick J. Angiolillo, Amal Arachiche, Richard H. Aster, Tiziano Barbui, Stefania Basili, Elisabeth M. Battinelli, Anthony A. Bavry, Wolfgang Bergmeier, Gerald Bertrand, Deepak L. Bhatt, Thomas A. Blair, Kamila Bledzka, Oliver Borst, Emma G. Bouck, Lawrence F. Brass, Paul F. Bray, Carol Briggs, Tomasz Brzoska, James B. Bussel, Marco Cattaneo, Subarna Chakravorty, Noel C. Chan, Shruti Chaturvedi, Beng H. Chong, Kenneth J. Clemetson, Jeannine M. Clemetson, Barry S. Coller, Gregory J. del Zoppo, Jenny M. Despotovic, Scott L. Diamond, J. Donald Easton, Koji Eto, Hervé Falet, Francisca Ferrer-Marin, Guido Finazzi, Robert Flaumenhaft, Jane E. Freedman, Andrew L. Frelinger, Kathleen Freson, Aleksandra Gasecka, Meinrad Gawaz, Silvia Giannini, Andreas Greinacher, Thomas Gremmel, Paul A. Gurbel, Elizabeth J. Haining, Xu Han, Paul Harrison, Catherine P.M. Hayward, Karin Hoffmeister, Anne-Mette Hvas, Sara J. Israels, Joseph E. Italiano, Young-Hoon Jeong, Andrew D. Johnson, Cecile Kaplan, Peter Karagiannis, Gregory J. Kato, Samuel Kemble, Kumaran Kolandaivelu, Milka Koupenova, David J. Kuter, Michele P. Lambert, Robert H. Lee, Jack Levin, Renhao Li, Zhenyu Li, Zihai Li, Anqi Li, Rossella Liani, Marie Lordkipanidzé, Viola Lorenz, Kellie R. Machlus, Dhruv Mahtta, Pier Mannuccio Mannucci, Keith R. McCrae, Alessandra Metelli, Alan D. Michelson, Karen A. Moffat, Jae Youn Moon, James H. Morrissey, Nicola J. Mutch, Zoltan Nagy, Heyu Ni, Phillip L.R. Nicolson, Marvin T. Nieman, Rienk Nieuwland, Marie-Blanche Onselaer, Carlo Patrono, Edward F. Plow, Mortimer Poncz, Man-Chiu Poon, Natalie S. Poulter, Izmarie Poventud-Fuentes, Patrick Provost, Jun Qin, Julie Rayes, Alexander P. Reiner, Brian Riesenberg, Irene A.G. Roberts, Matthew T. Rondina, Jesse W. Rowley, Francesca Santilli, Rüdiger E. Scharf, Yotis A. Senis, Anish Sharda, Alexa J. Siddon, Pia R.-M. Siljander, Pierluigi Tricoci, Paola Simeone, Stephanie A. Smith, Susan S. Smyth, Edward L. Snyder, Martha Sola-Visner, Timothy J. Stalker, Lucia Stefanini, Naoshi Sugimoto, Prithu Sundd, Udaya S. Tantry, Ayalew Tefferi, Steven G. Thomas, Mark R. Thomas, Maurizio Tomaiuolo, Christopher A. Tormey, Han-Mou Tsai, Francesco Violi, Theodore E. Warkentin, Steve P. Watson, Jeffrey I. Weitz, John Welsh, Andrew S. Weyrich, David A. Wilcox, Bill X. Wu, Michael R. Yeaman, Li Zhu, Guy A. Zimmerman, and Elizabeth R. Zunica
- Published
- 2019
- Full Text
- View/download PDF
42. Megakaryocyte Development and Platelet Formation
- Author
-
Kellie R. Machlus and Joseph E. Italiano
- Published
- 2019
- Full Text
- View/download PDF
43. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets
- Author
-
Allyn Morris, Pierre Cunin, Rim Bouslama, Peter A. Nigrovic, Andrew S. Weyrich, Li Guo, Joseph E. Italiano, Marta Martínez-Bonet, Pui Y. Lee, Nathan Nelson-Maney, Kellie R. Machlus, Eric Boilard, and Martha Sola-Visner
- Subjects
0303 health sciences ,Chemistry ,Vesicle ,Inflammation ,Cell biology ,Emperipolesis ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,Ezrin ,medicine.anatomical_structure ,Megakaryocyte ,Cytoplasm ,030220 oncology & carcinogenesis ,medicine ,Platelet ,medicine.symptom ,030304 developmental biology - Abstract
SummaryBone marrow megakaryocytes engulf neutrophils in a phenomenon termed emperipolesis. We show here that emperipolesis is a dynamic process mediated actively by both lineages, in part through the β2-integrin/ICAM-1/ezrin pathway. Tethered neutrophils enter in membrane-bound vesicles before penetrating into the megakaryocyte cytoplasm. Intracytoplasmic neutrophils develop membrane contiguity with the demarcation membrane system, thereby transferring membrane to the megakaryocyte and to daughter platelets. This phenomenon occurs in otherwise unmanipulated marrowin vivo, resulting in circulating platelets that bear membrane from non-megakaryocytic hematopoietic donors. Transit through megakaryocytes can be completed as rapidly as minutes, after which neutrophils egress intact. Emperipolesis is amplified in models of inflammation associated with platelet overproduction, contributing to platelet productionin vitroandin vivo.These findings identify emperipolesis as a new cell-in-cell interaction that enables neutrophils and potentially other cells passing through the megakaryocyte cytoplasm to modulate the production and membrane content of platelets.
- Published
- 2018
- Full Text
- View/download PDF
44. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets
- Author
-
Andrew S. Weyrich, Li Guo, Allyn Morris, Pierre Cunin, Rim Bouslama, Peter A. Nigrovic, Alexandra Wactor, Marta Martínez-Bonet, Pui Y. Lee, Joseph E. Italiano, Eric Boilard, Kellie R. Machlus, Nathan Nelson-Maney, and Martha Sola-Visner
- Subjects
0301 basic medicine ,Cytoplasm ,Mouse ,Neutrophils ,Cell Communication ,030204 cardiovascular system & hematology ,megakaryocyte ,Mice ,0302 clinical medicine ,Ezrin ,Megakaryocyte ,Platelet ,Biology (General) ,membrane ,platelet ,Chemistry ,General Neuroscience ,Vesicle ,neutrophil ,General Medicine ,Intercellular Adhesion Molecule-1 ,Cell biology ,Emperipolesis ,Haematopoiesis ,medicine.anatomical_structure ,Medicine ,medicine.symptom ,Megakaryocytes ,Research Article ,Blood Platelets ,QH301-705.5 ,Science ,Inflammation ,Bone Marrow Cells ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,General Immunology and Microbiology ,Membrane Transport Proteins ,Cell Biology ,Cytoskeletal Proteins ,030104 developmental biology ,CD18 Antigens - Abstract
Bone marrow megakaryocytes engulf neutrophils in a phenomenon termed emperipolesis. We show here that emperipolesis is a dynamic process mediated actively by both lineages, in part through the β2-integrin/ICAM-1/ezrin pathway. Tethered neutrophils enter in membrane-bound vesicles before penetrating into the megakaryocyte cytoplasm. Intracytoplasmic neutrophils develop membrane contiguity with the demarcation membrane system, thereby transferring membrane to the megakaryocyte and to daughter platelets. This phenomenon occurs in otherwise unmanipulated murine marrow in vivo, resulting in circulating platelets that bear membrane from non-megakaryocytic hematopoietic donors. Transit through megakaryocytes can be completed as rapidly as minutes, after which neutrophils egress intact. Emperipolesis is amplified in models of murine inflammation associated with platelet overproduction, contributing to platelet production in vitro and in vivo. These findings identify emperipolesis as a new cell-in-cell interaction that enables neutrophils and potentially other cells passing through the megakaryocyte cytoplasm to modulate the production and membrane content of platelets.
- Published
- 2018
45. Aspirin inhibits platelets from reprogramming breast tumor cells and promoting metastasis
- Author
-
Kelly E. Johnson, Wendy Y. Chen, Jodi A. Forward, Elisabeth M. Battinelli, Saleh El-Husayni, Julia R. Ceglowski, Mason D. Tippy, Michael W. Malloy, Kellie R. Machlus, Rajesh Kulenthirarajan, Joseph E. Italiano, and Harvey G. Roweth
- Subjects
0301 basic medicine ,Blood Platelets ,Breast Neoplasms ,Metastasis ,Thrombosis and Hemostasis ,Neovascularization ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,Medicine ,Humans ,Platelet ,Interleukin 8 ,Neoplasm Metastasis ,skin and connective tissue diseases ,PI3K/AKT/mTOR pathway ,Neoplasm Staging ,Tumor microenvironment ,Aspirin ,Neovascularization, Pathologic ,business.industry ,Hematology ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Cytokines ,Female ,medicine.symptom ,Signal transduction ,business ,Proto-Oncogene Proteins c-akt ,Platelet Aggregation Inhibitors ,Signal Transduction - Abstract
It is now recognized that compounds released from tumor cells can activate platelets, causing the release of platelet-derived factors into the tumor microenvironment. Several of these factors have been shown to directly promote neovascularization and metastasis, yet how the feedback between platelet releasate and the tumor cell affects metastatic phenotype remains largely unstudied. Here, we identify that breast tumor cells secrete high levels of interleukin 8 (IL-8, CXCL8) in response to platelet releasate, which promotes their invasive capacity. Furthermore, we found that platelets activate the Akt pathway in breast tumor cells, and inhibition of this pathway eliminated IL-8 production. We therefore hypothesized inhibiting platelets with aspirin could reverse the prometastatic effects of platelets on tumor cell signaling. Platelets treated with aspirin did not activate the Akt pathway, resulting in reduced IL-8 secretion and impaired tumor cell invasion. Of note, patients with breast cancer receiving aspirin had lower circulating IL-8, and their platelets did not increase tumor cell invasion compared with patients not receiving aspirin. Our data suggest platelets support breast tumor metastasis by inducing tumor cells to secrete IL-8. Our data further support that aspirin acts as an anticancer agent by disrupting the communication between platelets and breast tumor cells.
- Published
- 2018
46. In vitro Culture of Murine Megakaryocytes from Fetal Liver-derived Hematopoietic Stem Cells
- Author
-
Prakrith Vijey, Benjamin Posorske, and Kellie R. Machlus
- Subjects
0301 basic medicine ,Fetus ,urogenital system ,Chemistry ,Hematology ,General Medicine ,In Vitro Techniques ,Hematopoietic Stem Cells ,In vitro ,Article ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Megakaryocyte ,Precursor cell ,medicine ,Humans ,Platelet ,Stem cell ,Megakaryocytes - Abstract
Megakaryocytes (MKs) are specialized precursor cells committed to producing and proliferating platelets. In a cytoskeletal-driven process, mature MKs generate platelets by releasing thin cytoplasmic extensions, named proplatelets, into the sinusoids. Due to knowledge gaps in this process and mounting clinical demand for non-donor-based platelet sources, investigators are successfully developing artificial culture systems to recreate the environment of platelet biogenesis. Nevertheless, drawbacks in current methods entail elaborate procedures for stem cell enrichment, extensive growth periods, low MK yield, and poor proplatelet production. We propose a simple, robust method of primary MK culture that utilizes fetal livers from pregnant mice. Our technique reduces expansion time to 4 days, and generates ~15,000-20,000 MKs per liver. Approximately, 20-50% of these MKs produce structurally dense, high-quality proplatelets. In this review, we outline our method of MK culture and isolation.
- Published
- 2018
47. Megakaryocyte modification of platelets in thrombocytopenia
- Author
-
Harvey G. Roweth, Kellie R. Machlus, and Somayje Parvin
- Subjects
0301 basic medicine ,Blood Platelets ,Megakaryocyte differentiation ,Hemorrhage ,Thrombopoiesis ,03 medical and health sciences ,0302 clinical medicine ,Megakaryocyte ,Precursor cell ,Platelet production ,Medicine ,Animals ,Humans ,Platelet ,Cytoskeleton ,Whole blood ,Megakaryopoiesis ,Hemostasis ,business.industry ,Platelet Count ,Hematology ,Thrombocytopenia ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,business ,Megakaryocytes ,030215 immunology - Abstract
PURPOSE OF REVIEW Platelets are small, anucleate cells that circulate within the blood and play essential roles in preserving vascular integrity. However, abnormalities in either platelet production or destruction can result in thrombocytopenia, clinically defined by a platelet count lower than 150 000/μL of whole blood. Thrombocytopenia is frequently associated with impaired hemostatic responses to vascular injury and can be life-threatening because of bleeding complications. Megakaryocytes are the precursor cells responsible for platelet production, a process commonly referred to as thrombopoiesis. This review specifically discusses how perturbation of molecular mechanisms governing megakaryocyte differentiation and development manifest in various forms of thrombocytopenia. RECENT FINDINGS This review highlights the identification of novel transcriptional regulators of megakaryocyte maturation and platelet production. We also provide an update into the essential role of cytoskeletal regulation in thrombopoiesis, and how both megakaryopoiesis and platelet production are altered by anticancer therapeutics. Lastly, we focus on recent investigative approaches to treat thrombocytopenia and discuss future prospects in the field of megakaryocyte research. SUMMARY In patients where thrombocytopenia is not due to heightened platelet destruction or clearance, defects in megakaryocyte development should be considered.
- Published
- 2018
48. Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration
- Author
-
Paul R. Fortin, Tania Lévesque, Bernhard Nieswandt, Huiying Zhi, Matthew T. Rondina, Benoit Mailhot, Steve Lacroix, Nicolas Tessandier, Matthew J. Flick, Nathalie Cloutier, Anne Zufferey, Waliul I. Khan, Geneviève Marcoux, Francine Côté, Joseph E. Italiano, Isabelle Allaeys, Imene Melki, Louis Flamand, Steven E. McKenzie, Peter J. Newman, Guy G. Poirier, Kellie R. Machlus, Yann Becker, Eric Boilard, CHU de Québec–Université Laval, and Université Laval [Québec] (ULaval)
- Subjects
Adult ,Blood Platelets ,Male ,0301 basic medicine ,Serotonin ,Inflammation ,Antigen-Antibody Complex ,Platelet Glycoprotein GPIIb-IIIa Complex ,Sepsis ,Mice ,Young Adult ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Platelet ,Platelet activation ,Anaphylaxis ,ComputingMilieux_MISCELLANEOUS ,Mice, Knockout ,Multidisciplinary ,Platelet Count ,business.industry ,Receptors, IgG ,Platelet Activation ,Acquired immune system ,medicine.disease ,Shock, Septic ,Immune complex ,3. Good health ,Mice, Inbred C57BL ,030104 developmental biology ,PNAS Plus ,Shock (circulatory) ,Immunology ,Female ,medicine.symptom ,business - Abstract
There is a growing appreciation for the contribution of platelets to immunity; however, our knowledge mostly relies on platelet functions associated with vascular injury and the prevention of bleeding. Circulating immune complexes (ICs) contribute to both chronic and acute inflammation in a multitude of clinical conditions. Herein, we scrutinized platelet responses to systemic ICs in the absence of tissue and endothelial wall injury. Platelet activation by circulating ICs through a mechanism requiring expression of platelet Fcγ receptor IIA resulted in the induction of systemic shock. IC-driven shock was dependent on release of serotonin from platelet-dense granules secondary to platelet outside-in signaling by αIIbβ3 and its ligand fibrinogen. While activated platelets sequestered in the lungs and leaky vasculature of the blood-brain barrier, platelets also sequestered in the absence of shock in mice lacking peripheral serotonin. Unexpectedly, platelets returned to the blood circulation with emptied granules and were thereby ineffective at promoting subsequent systemic shock, although they still underwent sequestration. We propose that in response to circulating ICs, platelets are a crucial mediator of the inflammatory response highly relevant to sepsis, viremia, and anaphylaxis. In addition, platelets recirculate after degranulation and sequestration, demonstrating that in adaptive immunity implicating antibody responses, activated platelets are longer lived than anticipated and may explain platelet count fluctuations in IC-driven diseases.
- Published
- 2018
- Full Text
- View/download PDF
49. Teamwork makes the dream work in thrombopoiesis
- Author
-
Nathan L Asquith and Kellie R. Machlus
- Subjects
0301 basic medicine ,Platelet disorder ,Immunology ,Biology ,Biochemistry ,Thrombopoiesis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,LYL1 ,Megakaryocyte ,hemic and lymphatic diseases ,medicine ,Animals ,Transcription factor ,fungi ,GATA1 ,Cell Biology ,Hematology ,030104 developmental biology ,medicine.anatomical_structure ,RUNX1 ,chemistry ,FLI1 ,Cancer research ,030215 immunology - Abstract
In this issue of Blood, Chiu et al report the presence of functional redundancy between the transcription factors lymphoblastic leukemia 1 protein (LYL1) and stem cell leukemia protein (SCL) in the process of megakaryocyte (MK) maturation and platelet production in mice.1 This discovery may provide an answer to the long-standing mystery of why mutations in SCL protein do not present with any phenotypic platelet disorder, whereas mutations in its binding partner proteins GATA1, FLI1, RUNX1, and GFI1B cause hereditary thrombocytopenia.
- Published
- 2019
- Full Text
- View/download PDF
50. Deletion of the Arp2/3 complex in megakaryocytes leads to microthrombocytopenia in mice
- Author
-
Robert A. Campbell, Robert H. Lee, James E. Bear, Dorsaf Ghalloussi, Andrew S. Weyrich, Raymond Piatt, Jeremy D. Rotty, Swetha Pasala, Brian C. Cooley, Congying Wu, Kathryn O. Poe, David S. Paul, Wolfgang Bergmeier, Kellie R. Machlus, Joseph E. Italiano, and Caterina Casari
- Subjects
0301 basic medicine ,biology ,Integrin ,Arp2/3 complex ,Hematology ,macromolecular substances ,Actin cytoskeleton ,Cell biology ,Thrombosis and Hemostasis ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Actin filament branching ,Megakaryocyte ,medicine ,Cancer research ,biology.protein ,Platelet ,Platelet activation ,biological phenomena, cell phenomena, and immunity ,Actin - Abstract
Actin reorganization regulates key processes in platelet activation. Here we examined the role of the Arp2/3 complex, an essential component in actin filament branching, in platelet function. The Arpc2 gene, encoding the p34 subunit of the Arp2/3 complex, was deleted in the megakaryocyte lineage ( Arpc2 fl/fl PF4-Cre ). Deletion of the Arp2/3 complex resulted in marked microthrombocytopenia in mice, caused by premature platelet release into the bone marrow compartment and impaired platelet survival in circulation. Arpc2 fl/fl PF4-Cre platelets exhibited alterations in their actin cytoskeleton and their peripheral microtubule coil. Thrombocytopenia was alleviated following clodronate liposome-induced macrophage depletion in Arpc2 fl/fl PF4-Cre mice. Arpc2 fl/fl PF4-Cre platelets failed to spread and showed a mild defect in integrin activation and aggregation; however, no significant differences in hemostasis or thrombosis were observed between Arpc2 fl/fl PF4-Cre and control mice. Thus, Arp2/3 is critical for platelet homeostasis but plays only a minor role for vascular hemostasis.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.