126 results on '"Italiano JE"'
Search Results
2. Looking Under the Hood at the Cytoskeletal Engine of Platelet Production.
- Author
-
Italiano JE Jr, Payne C, and Bekendam RH
- Subjects
- Humans, Animals, Microtubules metabolism, Centrosome metabolism, Actins metabolism, Signal Transduction, Blood Platelets metabolism, Megakaryocytes metabolism, Megakaryocytes cytology, Thrombopoiesis physiology, Cytoskeleton metabolism
- Abstract
Blood platelets are anucleate cells essential for normal blood hemostasis. To maintain a normal platelet count of 150 000 to 400 000 per μL of blood, 10
11 platelets must be released each day from precursor cells called megakaryocytes. In this review, we aim to provide an overview of platelet production and evaluate the proposed mechanisms of platelet generation. We will discuss novel cytoskeletal mechanisms of platelet production, including microtubule and actin-based systems. We present new evidence that supports a cytoplasmic trigger for platelet production, discuss centrosome clustering as a new mechanism to trigger proplatelet production, and review new data supporting the bone marrow as the major location of platelet production., Competing Interests: J.E. Italiano Jr has a financial interest in and is a founder of Stellular Bio and Spry Bio. The interests of J.E. Italiano Jr are managed by Boston Children’s Hospital. The other authors report no conflicts.- Published
- 2025
- Full Text
- View/download PDF
3. Inhibition of RhoA-mediated secretory autophagy in megakaryocytes mitigates myelofibrosis in mice.
- Author
-
Becker IC, Barrachina MN, Lykins J, Camacho V, Stone AP, Chua BA, Signer RAJ, Machlus KR, Whiteheart SW, Roweth HG, and Italiano JE
- Abstract
Megakaryocytes (MKs) are large, polyploid cells that contribute to bone marrow homeostasis through the secretion of cytokines such as transforming growth factor β1 (TGFβ1). During neoplastic transformation, immature MKs accumulate in the bone marrow where they induce fibrotic remodeling ultimately resulting in myelofibrosis. Current treatment strategies aim to prevent MK hyperproliferation, however, little is understood about the potential of targeting dysregulated cytokine secretion from neoplastic MKs as a novel therapeutic avenue. Unconventional secretion of TGFβ1 as well as interleukin 1β (IL1β) via secretory autophagy occurs in cells other than MKs, which prompted us to investigate whether similar mechanisms are utilized by MKs. Here, we identified that TGFβ1 strongly co-localized with the autophagy marker light chain 3B in native MKs. Disrupting secretory autophagy by inhibiting the small GTPase RhoA or its downstream effector Rho kinase (ROCK) markedly reduced TGFβ1 and IL1β secretion in vitro . In vivo , conditional deletion of the essential autophagy gene Atg5 from the hematopoietic system limited megakaryocytosis and aberrant cytokine secretion in an MPL
W515L -driven transplant model. Similarly, mice with a selective deletion of Rhoa from the MK and platelet lineage were protected from progressive fibrosis. Finally, disease hallmarks in MPLW515L -transplanted mice were attenuated upon treatment with the autophagy inhibitor hydroxychloroquine or the ROCK inhibitor Y27632, either as monotherapy or in combination with the JAK2 inhibitor ruxolitinib. Overall, our data indicate that aberrant cytokine secretion is dependent on secretory autophagy downstream of RhoA, targeting of which represents a novel therapeutic avenue in the treatment of myelofibrosis., One Sentence Summary: TGFβ1 is released from megakaryocytes via RhoA-mediated secretory autophagy, and targeting this process can alleviate fibrosis progression in a preclinical mouse model of myelofibrosis.- Published
- 2024
- Full Text
- View/download PDF
4. CpG oligonucleotides induce acute murine thrombocytopenia dependent on toll-like receptor 9 and spleen tyrosine kinase pathways.
- Author
-
Johansson K, Maouia A, Rebetz J, Marcoux G, Shannon O, Italiano JE Jr, Narayanan P, Henry S, Shen L, and Semple JW
- Subjects
- Animals, Female, Humans, Mice, Acute Disease, Chemokine CCL2 metabolism, Chemokine CCL2 genetics, Disease Models, Animal, Mice, Inbred BALB C, Platelet Count, Protein Kinase Inhibitors pharmacology, THP-1 Cells, Blood Platelets metabolism, Blood Platelets drug effects, Blood Platelets enzymology, Immunoglobulins, Intravenous, Oligodeoxyribonucleotides pharmacology, Signal Transduction, Syk Kinase antagonists & inhibitors, Syk Kinase metabolism, Thrombocytopenia chemically induced, Thrombocytopenia blood, Toll-Like Receptor 9 metabolism
- Abstract
Background: CpG oligonucleotides (ODNs) are synthetic single-stranded DNA sequences that act as immunostimulants. They have been increasingly used to treat several cancers; however, thrombocytopenia is a potential recognized side effect of some sequences., Objectives: We tested the ability of 2 CpG ODNs (ODN 2395 and ISIS 120704) to induce thrombocytopenia when administered to BALB/c mice and determined mechanisms associated with thrombocytopenia., Methods: BALB/c mice were prebled and then injected with titrated doses of CpG ODNs, and platelet counts were determined. The mice were treated with intravenous immunoglobulin (IVIg) or various inhibitors and antagonists of toll-like receptor 9 (TLR9) and spleen tyrosine kinase (Syk) to determine their effects on thrombocytopenia., Results: Compared with saline-treated mice or mice treated with 2'-O-methoxyethyl-modified antisense ODN, both ODN 2395 and ISIS 120704 induced acute dose-dependent thrombocytopenia within 3 and 24 hours, respectively. The thrombocytopenia was associated with significant increases in plasma monocyte chemoattractant protein 1. IVIg administration significantly rescued the CpG ODN-induced thrombocytopenia, as did treatment with either a Syk inhibitor or TLR9 antagonists. In vitro, CpG ODN could activate human platelets and this correlated significantly with enhanced IVIg- and Syk-dependent phagocytosis by THP-1 monocytes., Conclusion: These results suggest that CpG ODNs induce acute inflammatory-associated (IVIg-sensitive) thrombocytopenia that can be alleviated by Syk- or TLR9-blockade, and an IVIg- and Syk-dependent platelet clearance pathway appears primarily responsible for the thrombocytopenia., Competing Interests: Declaration of competing interests There are no competing interests to disclose., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Targeting cargo to an unconventional secretory system within megakaryocytes allows the release of transgenic proteins from platelets.
- Author
-
Asquith NL, Becker IC, Scimone MT, Boccia T, Camacho V, Barrachina MN, Guo S, Freire D, Machlus K, Schulman S, Flaumenhaft R, and Italiano JE
- Subjects
- Animals, Humans, Mice, Inbred C57BL, Protein Transport, Mice, Endoplasmic Reticulum metabolism, Secretory Vesicles metabolism, Transgenes, Blood Platelets metabolism, Megakaryocytes metabolism, Green Fluorescent Proteins metabolism, Green Fluorescent Proteins genetics, Protein Disulfide-Isomerases metabolism, Protein Disulfide-Isomerases genetics
- Abstract
Background: Platelets are essential for hemostasis and thrombosis and play vital roles during metastatic cancer progression and infection. Hallmarks of platelet function are activation, cytoskeletal rearrangements, and the degranulation of their cellular contents upon stimulation. While α-granules and dense granules are the most studied platelet secretory granules, the dense tubular system (DTS) also functions as a secretory system for vascular thiol isomerases. However, how DTS cargo is packaged and transported from megakaryocytes (MKs) to platelets is poorly understood., Objectives: To underpin the mechanisms responsible for DTS cargo transport and leverage those for therapeutic protein packaging into platelets., Methods: A retroviral expression system combined with immunofluorescence confocal microscopy was employed to track protein DTS cargo protein disulfide isomerase fused to enhanced green fluorescent protein (eGFP-PDI) during platelet production. Murine bone marrow transplantation models were used to determine the release of therapeutic proteins from platelets., Results: We demonstrated that the endoplasmic reticulum retrieval motif Lys-Asp-Glu-Leu (KDEL) located at the C-terminus of protein disulfide isomerase was essential for the regular transport of eGFP-PDI-containing granules. eGFP-PDI
ΔKDEL , in which the retrieval signal was deleted, was aberrantly packaged, and its expression was upregulated within clathrin-coated endosomes. Finally, we found that ectopic transgenic proteins, such as tissue factor pathway inhibitor and interleukin 2, can be packaged into MKs and proplatelets by adding a KDEL retrieval sequence., Conclusion: Our data corroborate the DTS as a noncanonical secretory system in platelets and demonstrate that in vitro-generated MKs and platelets may be used as a delivery system for transgenic proteins during cellular therapy., Competing Interests: Declaration of competing interests J.E.I. has a financial interest in and is a founder of Stellular Bio, a biotechnology company focused on making donor-independent platelet-like cells at scale, and Spry Bio. Boston Children’s Hospital manages the interests of J.E.I.. R.F. is a founder of and consultant for Platelet Diagnostics. Beth Israel Deaconess Medical Center manages the interests of R.F.. All other authors declare no competing financial interests., (Copyright © 2024 International Society on Thrombosis and Haemostasis. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. Shear Stress Promotes Remodeling of Platelet Glycosylation via Upregulation of Platelet Glycosidase Activity: One More Thing.
- Author
-
Roka-Moiia Y, Lewis S, Cleveland E, Italiano JE, and Slepian MJ
- Abstract
Background: Mechanical circulatory support (MCS) is a mainstay of therapy for advanced and end-stage heart failure. Accompanied by systemic anticoagulation, contemporary MCS has become less thrombogenic, with bleeding complications emerging as a major cause of readmission and 1-year mortality. Shear-mediated platelet dysfunction and thrombocytopenia of undefined etiology are primary drivers of MCS-related bleeding. Recently, it has been demonstrated that deprivation of platelet surface glycosylation is associated with the decline of hemostatic function, microvesiculation, and premature apoptosis. We test the hypothesis that shear stress induces remodeling of platelet surface glycosylation via upregulation of glycosidase activity, thus facilitating platelet count decline and intense microvesiculation., Methods: Human gel-filtered platelets were exposed to continuous shear stress in vitro. Platelets and platelet-derived microparticles (PDMPs) were quantified via flow cytometry using size standard fluorescent nanobeads. Platelet surface glycosylation and NEU1 expression were evaluated using lectin- or immune-staining and multicolor flow cytometry; lectin blotting was utilized to verify glycosylation of individual glycoproteins. Platelet neuraminidase, galactosidase, hexosaminidase, and mannosidase activities were quantified using 4-methylumbelliferone-based fluorogenic substrates., Results: We demonstrate that shear stress promotes selective remodeling of platelet glycosylation via downregulation of 2,6-sialylation, terminal galactose, and mannose, while 2,3-sialylation remains largely unchanged. Shear-mediated deglycosylation is partially attenuated by neuraminidase inhibitors, strongly suggesting the involvement of platelet neuraminidase in observed phenomena. Shear stress increases platelet NEU1 surface expression and potentiates generation of numerous NEU1+ PDMPs. Platelets exhibit high basal hexosaminidase and mannosidase activities; basal activities of platelet neuraminidase and galactosidase are rather low and are significantly upregulated by shear stress. Shear stress of increased magnitude and duration promotes an incremental decline of platelet count and immense microvesiculation, both being further exacerbated by neuraminidase and partially attenuated by neuraminidase inhibition., Conclusion: Our data indicate that shear stress accumulation, consistent with supraphysiologic conditions of device-supported circulation, promotes remodeling of platelet glycosylation via selective upregulation of platelet glycosidase activity. Shear-mediated platelet deglycosylation is associated with platelet count drop and increased microvesiculation, thus offering a direct link between deglycosylation and thrombocytopenia observed in device-supported patients. Based on our findings, we propose a panel of molecular markers to be used for reliable detection of shear-mediated platelet deglycosylation in MCS., Competing Interests: None declared., (Thieme. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. What It Takes To Be a Platelet: Evolving Concepts in Platelet Production.
- Author
-
Carminita E, Becker IC, and Italiano JE
- Subjects
- Humans, Animals, Blood Platelets metabolism, Megakaryocytes cytology, Megakaryocytes metabolism, Thrombopoiesis physiology
- Abstract
Platelets are among the most abundant cells within the circulation. Given that the platelet lifespan is 7 to 10 days in humans, a constant production of around 100 billion platelets per day is required. Platelet production from precursor cells called megakaryocytes is one of the most enigmatic processes in human biology. Although it has been studied for over a century, there is still controversy about the exact mechanisms leading to platelet release into circulation. The formation of proplatelet extensions from megakaryocytes into bone marrow sinusoids is the best-described mechanism explaining the origin of blood platelets. However, using powerful imaging techniques, several emerging studies have recently raised challenging questions in the field, suggesting that small platelet-sized structures called buds might also contribute to the circulating platelet pool. How and whether these structures differ from microvesicles or membrane blebs, which have previously been described to be released from megakaryocytes, is still a matter of discussion. In this review, we will summarize what the past and present have revealed about platelet production and whether mature blood platelets might emerge via different mechanisms., Competing Interests: J.E. Italiano has a financial interest in and is a founder of Stellular Bio and Spry Bio. The interests of J.E. Italiano are managed by Boston Children’s Hospital. The other authors report no conflicts.
- Published
- 2024
- Full Text
- View/download PDF
8. Evidence for a cytoplasmic proplatelet promoting factor that triggers platelet production.
- Author
-
Italiano JE Jr and Machlus KR
- Subjects
- Humans, Cytoplasm metabolism, Thrombopoiesis, Blood Platelets metabolism
- Published
- 2024
- Full Text
- View/download PDF
9. Cell cycle-dependent centrosome clustering precedes proplatelet formation.
- Author
-
Becker IC, Wilkie AR, Nikols E, Carminita E, Roweth HG, Tilburg J, Sciaudone AR, Noetzli LJ, Fatima F, Couldwell G, Ray A, Mogilner A, Machlus KR, and Italiano JE Jr
- Subjects
- Animals, Mice, Mice, Knockout, Humans, Mitosis, Centrosome metabolism, Megakaryocytes metabolism, Megakaryocytes cytology, Cell Cycle, Blood Platelets metabolism, Kinesins metabolism, Kinesins genetics
- Abstract
Platelet-producing megakaryocytes (MKs) primarily reside in the bone marrow, where they duplicate their DNA content with each cell cycle resulting in polyploid cells with an intricate demarcation membrane system. While key elements of the cytoskeletal reorganizations during proplatelet formation have been identified, what initiates the release of platelets into vessel sinusoids remains largely elusive. Using a cell cycle indicator, we observed a unique phenomenon, during which amplified centrosomes in MKs underwent clustering following mitosis, closely followed by proplatelet formation, which exclusively occurred in G
1 of interphase. Forced cell cycle arrest in G1 increased proplatelet formation not only in vitro but also in vivo following short-term starvation of mice. We identified that inhibition of the centrosomal protein kinesin family member C1 (KIFC1) impaired clustering and subsequent proplatelet formation, while KIFC1-deficient mice exhibited reduced platelet counts. In summary, we identified KIFC1- and cell cycle-mediated centrosome clustering as an important initiator of proplatelet formation from MKs.- Published
- 2024
- Full Text
- View/download PDF
10. Dynamic actin/septin network in megakaryocytes coordinates proplatelet elaboration.
- Author
-
Becker IC, Wilkie AR, Unger BA, Sciaudone AR, Fatima F, Tsai IT, Xu K, Machlus KR, and Italiano JE
- Subjects
- Animals, Mice, Septins, Blood Platelets, Actin Cytoskeleton, Actins, Megakaryocytes
- 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
- 2024
- Full Text
- View/download PDF
11. The bone marrow is the primary site of thrombopoiesis.
- Author
-
Asquith NL, Carminita E, Camacho V, Rodriguez-Romera A, Stegner D, Freire D, Becker IC, Machlus KR, Khan AO, and Italiano JE
- Subjects
- Mice, Humans, Animals, Blood Platelets, Megakaryocytes, Spleen, Bone Marrow, Thrombopoiesis genetics
- Abstract
Abstract: Megakaryocytes (MKs) generate thousands of platelets over their lifespan. The roles of platelets in infection and inflammation has guided an interest to the study of extramedullary thrombopoiesis and therefore MKs have been increasingly reported within the spleen and lung. However, the relative abundance of MKs in these organs compared to the bone marrow and the scale of their contribution to the platelet pool in a steady state remain controversial. We investigated the relative abundance of MKs in the adult murine bone marrow, spleen, and lung using whole-mount light-sheet and quantitative histological imaging, flow cytometry, intravital imaging, and an assessment of single-cell RNA sequencing (scRNA-seq) repositories. Flow cytometry revealed significantly higher numbers of hematopoietic stem and progenitor cells and MKs in the murine bone marrow than in spleens or perfused lungs. Two-photon intravital and light-sheet microscopy, as well as quantitative histological imaging, confirmed these findings. Moreover, ex vivo cultured MKs from the bone marrow subjected to static or microfluidic platelet production assays had a higher capacity for proplatelet formation than MKs from other organs. Analysis of previously published murine and human scRNA-seq data sets revealed that only a marginal fraction of MK-like cells can be found within the lung and most likely only marginally contribute to platelet production in the steady state., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
12. Thrombopoietin levels in sepsis and septic shock - a systematic review and meta-analysis.
- Author
-
Liu C, Görlich D, Lowell CA, Italiano JE, Rossaint J, Bender M, Zarbock A, and Margraf A
- Subjects
- Adult, Humans, Thrombopoietin, Shock, Septic, Sepsis
- Abstract
Objectives: Sepsis is a life-threatening condition implicating an inadequate activation of the immune system. Platelets act as modulators and contributors to immune processes. Indeed, altered platelet turnover, thrombotic events, and changes in thrombopoietin levels in systemic inflammation have been reported, but thrombopoietin-levels in sepsis and septic-shock have not yet been systematically evaluated. We therefore performed a meta-analysis of thrombopoietin (TPO)-levels in patients with sepsis., Methods: Two independent reviewers screened records and full-text articles for inclusion. Scientific databases were searched for studies examining thrombopoietin levels in adult sepsis and septic-shock patients until August 1st 2022., Results: Of 95 items screened, six studies met the inclusion criteria, including 598 subjects. Both sepsis and severe sepsis were associated with increased levels of thrombopoietin (sepsis vs. control: standardized mean difference 3.06, 95 % CI 1.35-4.77; Z=3.50, p=0.0005) (sepsis vs. severe sepsis: standardized mean difference -1.67, 95 % CI -2.46 to -0.88; Z=4.14, p<0.0001). TPO-levels did not show significant differences between severe sepsis and septic shock patients but differed between sepsis and inflammation-associated non-septic controls. Overall, high heterogeneity and low sample size could be noted., Conclusions: Concluding, increased levels of thrombopoietin appear to be present both in sepsis and severe sepsis with high heterogeneity but thrombopoietin does not allow to differentiate between severe sepsis and septic-shock. TPO may potentially serve to differentiate sepsis from non-septic trauma and/or tissue damage related (systemic) inflammation. Usage of different assays and high heterogeneity demand standardization of methods and further large multicenter trials., (© 2023 Walter de Gruyter GmbH, Berlin/Boston.)
- Published
- 2023
- Full Text
- View/download PDF
13. Sodium bicarbonate as a local adjunctive agent for limiting platelet activation, aggregation, and adhesion within cardiovascular therapeutic devices.
- Author
-
Ammann KR, Outridge CE, Roka-Moiia Y, Muslmani S, Ding J, Italiano JE, Tomat E, Corbett S, and Slepian MJ
- Subjects
- Humans, Thrombin metabolism, Phosphatidylserines metabolism, Platelet Activation, Platelet Aggregation, Blood Platelets, Heparin pharmacology, Sodium Bicarbonate pharmacology, Sodium Bicarbonate metabolism, Thrombosis drug therapy, Thrombosis prevention & control
- Abstract
Cardiovascular therapeutic devices (CTDs) remain limited by thrombotic adverse events. Current antithrombotic agents limit thrombosis partially, often adding to bleeding. The Impella® blood pump utilizes heparin in 5% dextrose (D5W) as an internal purge to limit thrombosis. While effective, exogenous heparin often complicates overall anticoagulation management, increasing bleeding tendency. Recent clinical studies suggest sodium bicarbonate (bicarb) may be an effective alternative to heparin for local anti-thrombosis. We examined the effect of sodium bicarbonate on human platelet morphology and function to better understand its translational utility. Human platelets were incubated (60:40) with D5W + 25 mEq/L, 50 mEq/L, or 100 mEq/L sodium bicarbonate versus D5W or D5W + Heparin 50 U/mL as controls. pH of platelet-bicarbonate solutions mixtures was measured. Platelet morphology was examined via transmission electron microscopy; activation assessed via P-selectin expression, phosphatidylserine exposure and thrombin generation; and aggregation with TRAP-6, calcium ionophore, ADP and collagen quantified; adhesion to glass measured via fluorescence microscopy. Sodium bicarbonate did not alter platelet morphology but did significantly inhibit activation, aggregation, and adhesion. Phosphatidylserine exposure and thrombin generation were both reduced in a concentration-dependent manner-between 26.6 ± 8.2% (p = 0.01) and 70.7 ± 5.6% (p < 0.0001); and 14.0 ± 6.2% (p = 0.15) and 41.7 ± 6.8% (p = 0.03), respectively, compared to D5W control. Platelet aggregation via all agonists was also reduced, particularly at higher concentrations of bicarb. Platelet adhesion to glass was similarly reduced, between 0.04 ± 0.03% (p = 0.61) and 0.11 ± 0.04% (p = 0.05). Sodium bicarbonate has direct, local, dose-dependent effects limiting platelet activation and adhesion. Our results highlight the potential utility of sodium bicarbonate as a locally acting agent to limit device thrombosis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36.
- Author
-
Barrachina MN, Pernes G, Becker IC, Allaeys I, Hirsch TI, Groeneveld DJ, Khan AO, Freire D, Guo K, Carminita E, Morgan PK, Collins TJC, Mellett NA, Wei Z, Almazni I, Italiano JE, Luyendyk J, Meikle PJ, Puder M, Morgan NV, Boilard E, Murphy AJ, and Machlus KR
- Subjects
- Animals, Humans, Mice, Knockout, Thrombocytopenia metabolism, Male, Mice, Inbred C57BL, Platelet Count, Cells, Cultured, Female, Mice, Lipidomics, Megakaryocyte Progenitor Cells metabolism, Megakaryocyte Progenitor Cells cytology, Blood Platelets metabolism, Thrombopoiesis physiology, CD36 Antigens metabolism, CD36 Antigens genetics, Phospholipids metabolism, Megakaryocytes metabolism, Megakaryocytes cytology, Fatty Acids, Unsaturated metabolism
- Abstract
Lipids contribute to hematopoiesis and membrane properties and dynamics; however, little is known about the role of lipids in megakaryopoiesis. Here we show that megakaryocyte progenitors, megakaryocytes and platelets present a unique lipidome progressively enriched in polyunsaturated fatty acid (PUFA)-containing phospholipids. In vitro, inhibition of both exogenous fatty acid functionalization and uptake as well as 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 low platelets. 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 viable target to modulate platelet counts., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
15. A Critical Role for ERO1α in Arterial Thrombosis and Ischemic Stroke.
- Author
-
Jha V, Xiong B, Kumari T, Brown G, Wang J, Kim K, Lee J, Asquith N, Gallagher J, Asherman L, Lambert T, Bai Y, Du X, Min JK, Sah R, Javaheri A, Razani B, Lee JM, Italiano JE, and Cho J
- Subjects
- Animals, Mice, Blood Platelets metabolism, Calcium Signaling, Disulfides, Platelet Activation, Ischemic Stroke metabolism, Thrombosis
- Abstract
Background: Platelet adhesion and aggregation play a crucial role in arterial thrombosis and ischemic stroke. Here, we identify platelet ERO1α (endoplasmic reticulum oxidoreductase 1α) as a novel regulator of Ca
2+ signaling and a potential pharmacological target for treating thrombotic diseases., Methods: Intravital microscopy, animal disease models, and a wide range of cell biological studies were utilized to demonstrate the pathophysiological role of ERO1α in arteriolar and arterial thrombosis and to prove the importance of platelet ERO1α in platelet activation and aggregation. Mass spectrometry, electron microscopy, and biochemical studies were used to investigate the molecular mechanism. We used novel blocking antibodies and small-molecule inhibitors to study whether ERO1α can be targeted to attenuate thrombotic conditions., Results: Megakaryocyte-specific or global deletion of Ero1α in mice similarly reduced platelet thrombus formation in arteriolar and arterial thrombosis without affecting tail bleeding times and blood loss following vascular injury. We observed that platelet ERO1α localized exclusively in the dense tubular system and promoted Ca2+ mobilization, platelet activation, and aggregation. Platelet ERO1α directly interacted with STIM1 (stromal interaction molecule 1) and SERCA2 (sarco/endoplasmic reticulum Ca2+ -ATPase 2) and regulated their functions. Such interactions were impaired in mutant STIM1-Cys49/56Ser and mutant SERCA2-Cys875/887Ser. We found that ERO1α modified an allosteric Cys49-Cys56 disulfide bond in STIM1 and a Cys875-Cys887 disulfide bond in SERCA2, contributing to Ca2+ store content and increasing cytosolic Ca2+ levels during platelet activation. Inhibition of Ero1α with small-molecule inhibitors but not blocking antibodies attenuated arteriolar and arterial thrombosis and reduced infarct volume following focal brain ischemia in mice., Conclusions: Our results suggest that ERO1α acts as a thiol oxidase for Ca2+ signaling molecules, STIM1 and SERCA2, and enhances cytosolic Ca2+ levels, promoting platelet activation and aggregation. Our study provides evidence that ERO1α may be a potential target to reduce thrombotic events., Competing Interests: Disclosures None.- Published
- 2023
- Full Text
- View/download PDF
16. Spatial transcriptomics of murine bone marrow megakaryocytes at single-cell resolution.
- Author
-
Tilburg J, Stone AP, Billingsley JM, Scoville DK, Pavenko A, Liang Y, Italiano JE Jr, and Machlus KR
- Abstract
Background: While megakaryocytes are known for making platelets, recent single-cell RNA sequencing data have revealed subpopulations of megakaryocytes with predicted immunoregulatory and bone marrow niche-supporting roles. Although these studies uncovered interesting information regarding the transcriptional variation of megakaryocytes, the generation, localization, and regulation of these subsets have not yet been studied and therefore remain incompletely understood. Considering the complex organization of the bone marrow, we reasoned that the application of spatial transcriptomic approaches could help dissect megakaryocyte heterogeneity within a spatiotemporal context., Objectives: The aim of this study was to combine spatial context and transcriptomics to assess the heterogeneity of murine bone marrow megakaryocytes in situ at a single-cell level., Methods: Bone marrow sections were obtained from femurs of C57BL/6J mice. Using the murine whole transcriptome array on the Nanostring GeoMx digital spatial profiling platform, we profiled 44 individual megakaryocytes (CD41
+ by immunofluorescence) in situ throughout the bone marrow, both adjacent and nonadjacent to the endothelium (directly in contact with vascular endothelial-cadherin-positive cells)., Results: Principal component analysis revealed no association between transcriptomic profile and adjacency to the vasculature. However, there was a significant effect of proximal vs distal regions of the bone. Two and 3 genes were found overexpressed in the proximal and distal sides, respectively. Of note, proplatelet basic protein and platelet factor 4, 2 genes associated with platelet production, had higher expression in proximal megakaryocytes ., Conclusion: This study indicates a possible effect of spatial location on megakaryocyte heterogeneity and substantiate further interest in investigating megakaryocyte subpopulations in the context of their spatial orientation., (© 2023 The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
17. Shear-Mediated Platelet Microparticles Demonstrate Phenotypic Heterogeneity as to Morphology, Receptor Distribution, and Hemostatic Function.
- Author
-
Roka-Moiia Y, Ammann KR, Miller-Gutierrez S, Sheriff J, Bluestein D, Italiano JE, Flaumenhaft RC, and Slepian MJ
- Subjects
- Humans, Thrombin metabolism, Blood Platelets metabolism, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Platelet Activation, Stress, Mechanical, Cell-Derived Microparticles metabolism, Hemostatics metabolism
- Abstract
Implantable Cardiovascular Therapeutic Devices (CTD), while lifesaving, impart supraphysiologic shear stress to platelets, resulting in thrombotic and bleeding coagulopathy. We previously demonstrated that shear-mediated platelet dysfunction is associated with downregulation of platelet GPIb-IX-V and αIIbβ3 receptors via generation of Platelet-Derived MicroParticles (PDMPs). Here, we test the hypothesis that sheared PDMPs manifest phenotypical heterogeneity of morphology and receptor surface expression and modulate platelet hemostatic function. Human gel-filtered platelets were exposed to continuous shear stress. Alterations of platelet morphology were visualized using transmission electron microscopy. Surface expression of platelet receptors and PDMP generation were quantified by flow cytometry. Thrombin generation was quantified spectrophotometrically, and platelet aggregation was measured by optical aggregometry. Shear stress promotes notable alterations in platelet morphology and ejection of distinctive types of PDMPs. Shear-mediated microvesiculation is associated with the remodeling of platelet receptors, with PDMPs expressing significantly higher levels of adhesion receptors (α
IIb β3 , GPIX, PECAM-1, P-selectin, and PSGL-1) and agonist receptors (P2 Y12 and PAR1). Sheared PDMPs promote thrombin generation and inhibit platelet aggregation induced by collagen and ADP. Sheared PDMPs demonstrate phenotypic heterogeneity as to morphology and defined patterns of surface receptors and impose a bidirectional effect on platelet hemostatic function. PDMP heterogeneity suggests that a range of mechanisms are operative in the microvesiculation process, contributing to CTD coagulopathy and posing opportunities for therapeutic manipulation.- Published
- 2023
- Full Text
- View/download PDF
18. Proceedings of the immune thrombocytopenia summit: new concepts in mechanisms, diagnosis, and management.
- Author
-
Mahamad S, Modi D, Al-Samkari H, Cuker A, Despotovic JM, Italiano JE, Lambert MP, Lee EJ, Rondina MT, Sholzberg M, Kruse C, Larché M, Nazy I, Miller MS, and Arnold DM
- Abstract
The inaugural McMaster Immune Thrombocytopenia (ITP) Summit was held virually in 2021. The objectives of the Summit were to recognize the difficulties in establishing the diagnosis of ITP and to understand gaps in current knowledge of ITP mechanisms that might lead to better diagnostic approaches and treatments. The half-day program consisted of virtual educational sessions targeting clinicians and basic scientists. The planning committee chose 8 topics to review that would cover current knowledge and inform future research priorities. In this report, we summarized the presentations delivered at the 2021 McMaster ITP Summit and the discussions. Based on the information presented at the Summit, the following research priorities were identified: 1) investigation of platelet production as a target for ITP treatments; 2) characterization of antigen processing and antigen presentation on platelets; 3) interaction between megakaryocytes and the immune system; 4) the role for ITP gene panels; 5) the need for better methods for platelet antibody testing; 6) the role of prediction models for diagnosis and prognosis; 7) new treatment strategies, including intensification of initial therapy; and 8) personalized treatment algorithms., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
19. Platelets upregulate tumor cell programmed death ligand 1 in an epidermal growth factor receptor-dependent manner in vitro.
- Author
-
Guo Q, Malloy MW, Roweth HG, McAllister SS, Italiano JE, and Battinelli EM
- Subjects
- United States, Humans, Epidermal Growth Factor pharmacology, Interferon-gamma pharmacology, Blood Platelets metabolism, Immune Checkpoint Inhibitors, Immune Checkpoint Proteins, Cetuximab, Platelet Aggregation Inhibitors, Ticagrelor, ErbB Receptors metabolism, Aspirin, Antibodies, Neutralizing, B7-H1 Antigen metabolism, Neoplasms drug therapy
- Abstract
Programmed death ligand 1 (PD-L1) is an immune checkpoint protein that suppresses cytotoxic T lymphocytes and is often overexpressed in cancers. Due to favorable clinical trial results, immune checkpoint inhibition (ICI) is part of Food and Drug Administration approved immuno-oncology therapies; however, not all patients benefit from ICI therapy. High blood platelet-to-lymphocyte ratio has been associated with failure of ICI treatment, but whether platelets have a role in hindering ICI response is unclear. Here, we report that coculturing platelets with cancer cell lines increased protein and gene expression of tumor cell PD-L1, which was reduced by antiplatelet agents, such as aspirin and ticagrelor. Platelet cytokine arrays revealed that the well-established cytokines, including interferon-γ, were not the main regulators of platelet-mediated PD-L1 upregulation. Instead, the high molecular weight epidermal growth factor (EGF) is abundant in platelets, which caused an upregulation of tumor cell PD-L1. Both an EGF-neutralizing antibody and cetuximab (EGF receptor [EGFR] monoclonal antibody) inhibited platelet-induced increases in tumor cell PD-L1, suggesting that platelets induce tumor cell PD-L1 in an EGFR-dependent manner. Our data reveal a novel mechanism for platelets in tumor immune escape and warrant further investigation to determine if targeting platelets improves ICI therapeutic responses., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Pro-inflammatory megakaryocyte gene expression in murine models of breast cancer.
- Author
-
Roweth HG, Malloy MW, Goreczny GJ, Becker IC, Guo Q, Mittendorf EA, Italiano JE Jr, McAllister SS, and Battinelli EM
- Subjects
- Animals, Blood Platelets metabolism, Cathepsin G metabolism, Disease Models, Animal, Gene Expression, Lipocalin-2 metabolism, Mice, Megakaryocytes, Neoplasms metabolism
- Abstract
Despite abundant research demonstrating that platelets can promote tumor cell metastasis, whether primary tumors affect platelet-producing megakaryocytes remains understudied. In this study, we used a spontaneous murine model of breast cancer to show that tumor burden reduced megakaryocyte number and size and disrupted polyploidization. Single-cell RNA sequencing demonstrated that megakaryocytes from tumor-bearing mice exhibit a pro-inflammatory phenotype, epitomized by increased Ctsg , Lcn2 , S100a8 , and S100a9 transcripts. Protein S100A8/A9 and lipocalin-2 levels were also increased in platelets, suggesting that tumor-induced alterations to megakaryocytes are passed on to their platelet progeny, which promoted in vitro tumor cell invasion and tumor cell lung colonization to a greater extent than platelets from wild-type animals. Our study is the first to demonstrate breast cancer-induced alterations in megakaryocytes, leading to qualitative changes in platelet content that may feedback to promote tumor metastasis.
- Published
- 2022
- Full Text
- View/download PDF
21. DNA Origami-Platelet Adducts: Nanoconstruct Binding without Platelet Activation.
- Author
-
Roka-Moiia Y, Walawalkar V, Liu Y, Italiano JE, Slepian MJ, and Taylor RE
- Subjects
- Blood Platelets, DNA metabolism, DNA Adducts, Humans, Platelet Activation, Cell-Derived Microparticles
- Abstract
Objective . Platelets are small, mechanosensitive blood cells responsible for maintaining vascular integrity and activatable on demand to limit bleeding and facilitate thrombosis. While circulating in the blood, platelets are exposed to a range of mechanical and chemical stimuli, with the platelet membrane being the primary interface and transducer of outside-in signaling. Sensing and modulating these interface signals would be useful to study mechanochemical interactions; yet, to date, no methods have been defined to attach adducts for sensor fabrication to platelets without triggering platelet activation. We hypothesized that DNA origami, and methods for its attachment, could be optimized to enable nonactivating instrumentation of the platelet membrane. Approach and Results . We designed and fabricated multivalent DNA origami nanotile constructs to investigate nanotile hybridization to membrane-embedded single-stranded DNA-tetraethylene glycol cholesteryl linkers. Two hybridization protocols were developed and validated (Methods I and II) for rendering high-density binding of DNA origami nanotiles to human platelets. Using quantitative flow cytometry, we showed that DNA origami binding efficacy was significantly improved when the number of binding overhangs was increased from two to six. However, no additional binding benefit was observed when increasing the number of nanotile overhangs further to 12. Using flow cytometry and transmission electron microscopy, we verified that hybridization with DNA origami constructs did not cause alterations in the platelet morphology, activation, aggregation, or generation of platelet-derived microparticles. Conclusions . Herein, we demonstrate that platelets can be successfully instrumented with DNA origami constructs with no or minimal effect on the platelet morphology and function. Our protocol allows for efficient high-density binding of DNA origami to platelets using low quantities of the DNA material to label a large number of platelets in a timely manner. Nonactivating platelet-nanotile adducts afford a path for advancing the development of DNA origami nanoconstructs for cell-adherent mechanosensing and therapeutic agent delivery.
- Published
- 2022
- Full Text
- View/download PDF
22. Don't you forget about me(gakaryocytes).
- Author
-
Tilburg J, Becker IC, and Italiano JE
- Subjects
- Blood Platelets metabolism, Bone Marrow, Hematopoietic Stem Cells, Megakaryocytes metabolism, Thrombopoiesis genetics
- Abstract
Platelets (small, anucleate cell fragments) derive from large precursor cells, megakaryocytes (MKs), that reside in the bone marrow. MKs emerge from hematopoietic stem cells in a complex differentiation process that involves cytoplasmic maturation, including the formation of the demarcation membrane system, and polyploidization. The main function of MKs is the generation of platelets, which predominantly occurs through the release of long, microtubule-rich proplatelets into vessel sinusoids. However, the idea of a 1-dimensional role of MKs as platelet precursors is currently being questioned because of advances in high-resolution microscopy and single-cell omics. On the one hand, recent findings suggest that proplatelet formation from bone marrow-derived MKs is not the only mechanism of platelet production, but that it may also occur through budding of the plasma membrane and in distant organs such as lung or liver. On the other hand, novel evidence suggests that MKs not only maintain physiological platelet levels but further contribute to bone marrow homeostasis through the release of extracellular vesicles or cytokines, such as transforming growth factor β1 or platelet factor 4. The notion of multitasking MKs was reinforced in recent studies by using single-cell RNA sequencing approaches on MKs derived from adult and fetal bone marrow and lungs, leading to the identification of different MK subsets that appeared to exhibit immunomodulatory or secretory roles. In the following article, novel insights into the mechanisms leading to proplatelet formation in vitro and in vivo will be reviewed and the hypothesis of MKs as immunoregulatory cells will be critically discussed., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
23. The triple crown of platelet generation.
- Author
-
Becker IC and Italiano JE
- Subjects
- Blood Platelets, Ticlopidine
- Published
- 2022
- Full Text
- View/download PDF
24. Sequence-specific 2'-O-methoxyethyl antisense oligonucleotides activate human platelets through glycoprotein VI, triggering formation of platelet-leukocyte aggregates.
- Author
-
Slingsby MHL, Vijey P, Tsai IT, Roweth H, Couldwell G, Wilkie AR, Gaus H, Goolsby JM, Okazaki R, Terkovich BE, Semple JW, Thon JN, Henry SP, Narayanan P, and Italiano JE Jr
- Subjects
- Humans, Leukocytes, Platelet Activation, Platelet Count, Blood Platelets, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense pharmacology, Oligonucleotides, Antisense therapeutic use
- Abstract
Antisense oligonucleotides (ASO) are DNA-based, disease-modifying drugs. Clinical trials with 2'-O-methoxyethyl (2'MOE) ASO have shown dose- and sequence-specific lowering of platelet counts according to two phenotypes. Phenotype 1 is a moderate (but not clinically severe) drop in platelet count. Phenotype 2 is rare, severe thrombocytopenia. This article focuses on the underlying cause of the more common phenotype 1, investigating the effects of ASO on platelet production and platelet function. Five phosphorothioate ASO were studied: three 2'MOE sequences; 487660 (no effects on platelet count), 104838 (associated with phenotype 1), and 501861 (effects unknown) and two CpG sequences; 120704 and ODN 2395 (known to activate platelets). Human cord bloodderived megakaryocytes were treated with these ASO to study their effects on proplatelet production. Platelet activation (determined by surface Pselectin) and platelet-leukocyte aggregates were analyzed in ASO-treated blood from healthy human volunteers. None of the ASO inhibited proplatelet production by human megakaryocytes. All the ASO were shown to bind to the platelet receptor glycoprotein VI (KD ~0.2-1.5 mM). CpG ASO had the highest affinity to glycoprotein VI, the most potent platelet-activating effects and led to the greatest formation of platelet-leukocyte aggregates. 2'MOE ASO 487660 had no detectable platelet effects, while 2'MOE ASOs 104838 and 501861 triggered moderate platelet activation and SYKdependent formation of platelet-leukocyte aggregates. Donors with higher platelet glycoprotein VI levels had greater ASO-induced platelet activation. Sequence-dependent ASO-induced platelet activation and platelet-leukocyte aggregates may explain phenotype 1 (moderate drops in platelet count). Platelet glycoprotein VI levels could be useful as a screening tool to identify patients at higher risk of ASO-induced platelet side effects.
- Published
- 2022
- Full Text
- View/download PDF
25. The secreted tyrosine kinase VLK is essential for normal platelet activation and thrombus formation.
- Author
-
Revollo L, Merrill-Skoloff G, De Ceunynck K, Dilks JR, Guo S, Bordoli MR, Peters CG, Noetzli L, Ionescu A, Rosen V, Italiano JE, Whitman M, and Flaumenhaft R
- Subjects
- Animals, Blood Platelets pathology, Gene Deletion, HEK293 Cells, Humans, Mice, Transgenic, Protein-Tyrosine Kinases genetics, Thrombosis pathology, Blood Platelets metabolism, Platelet Activation, Protein-Tyrosine Kinases metabolism, Thrombosis metabolism
- Abstract
Tyrosine phosphorylation of extracellular proteins is observed in cell cultures and in vivo, but little is known about the functional roles of tyrosine phosphorylation of extracellular proteins. Vertebrate lonesome kinase (VLK) is a broadly expressed secretory pathway tyrosine kinase present in platelet α-granules. It is released from platelets upon activation and phosphorylates substrates extracellularly. Its role in platelet function, however, has not been previously studied. In human platelets, we identified phosphorylated tyrosines mapped to luminal or extracellular domains of transmembrane and secreted proteins implicated in the regulation of platelet activation. To determine the role of VLK in extracellular tyrosine phosphorylation and platelet function, we generated mice with a megakaryocyte/platelet-specific deficiency of VLK. Platelets from these mice are normal in abundance and morphology but have significant changes in function both in vitro and in vivo. Resting and thrombin-stimulated VLK-deficient platelets exhibit a significant decrease in several tyrosine phosphobands. Results of functional testing of VLK-deficient platelets show decreased protease-activated receptor 4-mediated and collagen-mediated platelet aggregation but normal responses to adenosine 5'-diphosphate. Dense granule and α-granule release are reduced in these platelets. Furthermore, VLK-deficient platelets exhibit decreased protease-activated receptor 4-mediated Akt (S473) and Erk1/2 (T202/Y204) phosphorylation, indicating altered proximal signaling. In vivo, mice lacking VLK in megakaryocytes/platelets display strongly reduced platelet accumulation and fibrin formation after laser-induced injury of cremaster arterioles compared with control mice but with normal bleeding times. These studies show that the secretory pathway tyrosine kinase VLK is critical for stimulus-dependent platelet activation and thrombus formation, providing the first evidence that a secreted protein kinase is required for normal platelet function., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
26. Microvesicles, but not platelets, bud off from mouse bone marrow megakaryocytes.
- Author
-
Italiano JE, Bender M, Merrill-Skoloff G, Ghevaert C, Nieswandt B, and Flaumenhaft R
- Subjects
- Animals, Megakaryocytes cytology, Mice, Blood Platelets cytology, Bone Marrow ultrastructure, Cell-Derived Microparticles ultrastructure, Megakaryocytes ultrastructure
- Published
- 2021
- Full Text
- View/download PDF
27. Platelet Dysfunction During Mechanical Circulatory Support: Elevated Shear Stress Promotes Downregulation of α IIb β 3 and GPIb via Microparticle Shedding Decreasing Platelet Aggregability.
- Author
-
Roka-Moiia Y, Miller-Gutierrez S, Palomares DE, Italiano JE, Sheriff J, Bluestein D, and Slepian MJ
- Subjects
- Adenosine Diphosphate pharmacology, Blood Platelets drug effects, Cell-Derived Microparticles drug effects, Down-Regulation, Female, Humans, Male, Oligopeptides pharmacology, P-Selectin blood, Stress, Mechanical, Blood Platelets metabolism, Cell-Derived Microparticles metabolism, Mechanotransduction, Cellular, Platelet Aggregation drug effects, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Platelet Glycoprotein GPIb-IX Complex metabolism
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
28. VWF maturation and release are controlled by 2 regulators of Weibel-Palade body biogenesis: exocyst and BLOC-2.
- Author
-
Sharda AV, Barr AM, Harrison JA, Wilkie AR, Fang C, Mendez LM, Ghiran IC, Italiano JE, and Flaumenhaft R
- Subjects
- Endosomes metabolism, Human Umbilical Vein Endothelial Cells cytology, Humans, Limonins pharmacology, Exocytosis, Human Umbilical Vein Endothelial Cells metabolism, Intracellular Signaling Peptides and Proteins metabolism, Weibel-Palade Bodies metabolism, von Willebrand Factor metabolism
- Abstract
von Willebrand factor (VWF) is an essential hemostatic protein that is synthesized in endothelial cells and stored in Weibel-Palade bodies (WPBs). Understanding the mechanisms underlying WPB biogenesis and exocytosis could enable therapeutic modulation of endogenous VWF, yet optimal targets for modulating VWF release have not been established. Because biogenesis of lysosomal related organelle-2 (BLOC-2) functions in the biogenesis of platelet dense granules and melanosomes, which like WPBs are lysosome-related organelles, we hypothesized that BLOC-2-dependent endolysosomal trafficking is essential for WPB biogenesis and sought to identify BLOC-2-interacting proteins. Depletion of BLOC-2 caused misdirection of cargo-carrying transport tubules from endosomes, resulting in immature WPBs that lack endosomal input. Immunoprecipitation of BLOC-2 identified the exocyst complex as a binding partner. Depletion of the exocyst complex phenocopied BLOC-2 depletion, resulting in immature WPBs. Furthermore, releasates of immature WPBs from either BLOC-2 or exocyst-depleted endothelial cells lacked high-molecular weight (HMW) forms of VWF, demonstrating the importance of BLOC-2/exocyst-mediated endosomal input during VWF maturation. However, BLOC-2 and exocyst showed very different effects on VWF release. Although BLOC-2 depletion impaired exocytosis, exocyst depletion augmented WPB exocytosis, indicating that it acts as a clamp. Exposure of endothelial cells to a small molecule inhibitor of exocyst, Endosidin2, reversibly augmented secretion of mature WPBs containing HMW forms of VWF. These studies show that, although BLOC-2 and exocyst cooperate in WPB formation, only exocyst serves to clamp WPB release. Exocyst function in VWF maturation and release are separable, a feature that can be exploited to enhance VWF release., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
29. High-content, label-free analysis of proplatelet production from megakaryocytes.
- Author
-
French SL, Vijey P, Karhohs KW, Wilkie AR, Horin LJ, Ray A, Posorske B, Carpenter AE, Machlus KR, and Italiano JE Jr
- Subjects
- Blood Platelets, Cells, Cultured, Humans, Thrombopoiesis, Megakaryocytes, Thrombocytopenia
- Abstract
Background: 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 originating from the MK cell body., Objectives: Proplatelet formation is an asynchronous and dynamic process that poses unique challenges for researchers to accurately capture and analyze. We have designed an open-source, high-content, high-throughput, label-free analysis platform., Methods: 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 processed by a customized CellProfiler pipeline to identify and filter structures of interest based on morphology. A subsequent reinforcement classification, by CellProfiler Analyst, improves the detection of cellular structures., Results: This workflow yields the percent of proplatelet production, area, count of proplatelets and MKs, and 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., Conclusion: 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., (© 2020 International Society on Thrombosis and Haemostasis.)
- Published
- 2020
- Full Text
- View/download PDF
30. Platelet-derived extracellular vesicles infiltrate and modify the bone marrow during inflammation.
- Author
-
French SL, Butov KR, Allaeys I, Canas J, Morad G, Davenport P, Laroche A, Trubina NM, Italiano JE, Moses MA, Sola-Visner M, Boilard E, Panteleev MA, and Machlus KR
- Subjects
- Animals, Bone Marrow, Inflammation, Megakaryocytes, Mice, Blood Platelets, Extracellular Vesicles
- 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., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
31. Sniffing out the aroma(tase) of drug-induced thrombocytopenia.
- Author
-
Asquith NL and Italiano JE
- Subjects
- Aromatase, Humans, Immunomodulation, Odorants analysis, Pharmaceutical Preparations, Thrombocytopenia chemically induced
- Published
- 2020
- Full Text
- View/download PDF
32. Platelet Activation via Shear Stress Exposure Induces a Differing Pattern of Biomarkers of Activation versus Biochemical Agonists.
- Author
-
Roka-Moiia Y, Walk R, Palomares DE, Ammann KR, Dimasi A, Italiano JE, Sheriff J, Bluestein D, and Slepian MJ
- Subjects
- Apoptosis drug effects, Biomarkers blood, Blood Coagulation drug effects, Blood Platelets metabolism, Blood Platelets pathology, Caspase 3 blood, Humans, Membrane Potential, Mitochondrial drug effects, P-Selectin blood, Phosphatidylserines blood, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Stress, Mechanical, Blood Platelets drug effects, Calcium Signaling drug effects, Mechanotransduction, Cellular, Platelet Activation drug effects
- Abstract
Background: Implantable cardiovascular therapeutic devices, while hemodynamically effective, remain limited by thrombosis. A driver of device-associated thrombosis is shear-mediated platelet activation (SMPA). Underlying mechanisms of SMPA, as well as useful biomarkers able to detect and discriminate mechanical versus biochemical platelet activation, are poorly defined. We hypothesized that SMPA induces a differing pattern of biomarkers compared with biochemical agonists., Methods: Gel-filtered human platelets were subjected to mechanical activation via either uniform constant or dynamic shear; or to biochemical activation by adenosine diphosphate (ADP), thrombin receptor-activating peptide 6 (TRAP-6), thrombin, collagen, epinephrine, or arachidonic acid. Markers of platelet activation (P-selectin, integrin αIIbβ3 activation) and apoptosis (mitochondrial membrane potential, caspase 3 activation, and phosphatidylserine externalization [PSE]) were examined using flow cytometry. Platelet procoagulant activity was detected by chromogenic assay measuring thrombin generation. Contribution of platelet calcium flux in SMPA was tested employing calcium chelators, ethylenediaminetetraacetic acid (EDTA), and BAPTA-AM., Results: Platelet exposure to continuous shear stress, but not biochemical agonists, resulted in a dramatic increase of PSE and procoagulant activity, while no integrin αIIbβ3 activation occurred, and P-selectin levels remained barely elevated. SMPA was associated with dissipation of mitochondrial membrane potential, but no caspase 3 activation was observed. Shear-mediated PSE was significantly decreased by chelation of extracellular calcium with EDTA, while intracellular calcium depletion with BAPTA-AM had no significant effect. In contrast, biochemical agonists ADP, TRAP-6, arachidonic acid, and thrombin were potent inducers of αIIbβ3 activation and/or P-selectin exposure. This differing pattern of biomarkers seen for SMPA for continuous uniform shear was replicated in platelets exposed to dynamic shear stress via circulation through a ventricular assist device-propelled circulatory loop., Conclusion: Elevated shear stress, but not biochemical agonists, induces a differing pattern of platelet biomarkers-with enhanced PSE and thrombin generation on the platelet surface. This differential biomarker phenotype of SMPA offers the potential for early detection and discrimination from that mediated by biochemical agonists., Competing Interests: Y.R.-M., D.B., and M.J.S. have a patent PCT/US2019/037528 pending to University of Arizona.J. I. reports personal fees from Platelet BioGenesis, outside the submitted work., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2020
- Full Text
- View/download PDF
33. Platelet alloantibody detection: moving ahead.
- Author
-
Wilkie AR and Italiano JE
- Subjects
- Blood Platelets, Graft Rejection, Humans, Megakaryocytes, Induced Pluripotent Stem Cells, Isoantibodies
- Published
- 2019
- Full Text
- View/download PDF
34. Megakaryocytes package contents into separate α-granules that are differentially distributed in platelets.
- Author
-
Battinelli EM, Thon JN, Okazaki R, Peters CG, Vijey P, Wilkie AR, Noetzli LJ, Flaumenhaft R, and Italiano JE
- Subjects
- Animals, Biological Transport, Biomarkers, Cell Differentiation, Fluorescent Antibody Technique, Humans, Megakaryocytes cytology, Mice, Thrombopoiesis, Blood Platelets metabolism, Cytoplasmic Granules metabolism, Megakaryocytes metabolism
- Abstract
In addition to their primary roles in hemostasis and thrombosis, platelets participate in many other physiological and pathological processes, including, but not limited to inflammation, wound healing, tumor metastasis, and angiogenesis. Among their most interesting properties is the large number of bioactive proteins stored in their α-granules, the major storage granule of platelets. We previously showed that platelets differentially package pro- and antiangiogenic proteins in distinct α-granules that undergo differential release upon platelet activation. Nevertheless, how megakaryocytes achieve differential packaging is not fully understood. In this study, we use a mouse megakaryocyte culture system and endocytosis assay to establish when and where differential packaging occurs during platelet production. Live cell microscopy of primary mouse megakaryocytes incubated with fluorescently conjugated fibrinogen and endostatin showed differential endocytosis and packaging of the labeled proteins into distinct α-granule subpopulations. Super-resolution microscopy of mouse proplatelets and human whole-blood platelet α-granules simultaneously probed for 2 different membrane proteins (VAMP-3 and VAMP-8), and multiple granular content proteins (bFGF, ENDO, TSP, VEGF) confirmed differential packaging of protein contents into α-granules. These data suggest that megakaryocytes differentially sort and package α-granule contents, which are preserved as α-granule subpopulations during proplatelet extension and platelet production., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
35. Anti-apoptotic BCL2L2 increases megakaryocyte proplatelet formation in cultures of human cord blood.
- Author
-
Bhatlekar S, Basak I, Edelstein LC, Campbell RA, Lindsey CR, Italiano JE Jr, Weyrich AS, Rowley JW, Rondina MT, Sola-Visner M, and Bray PF
- Subjects
- Antigens, Differentiation biosynthesis, Cells, Cultured, Flow Cytometry, Gene Expression Regulation, Humans, Apoptosis Regulatory Proteins biosynthesis, Fetal Blood cytology, Fetal Blood metabolism, Megakaryocytes cytology, Megakaryocytes metabolism
- Abstract
Apoptosis is a recognized limitation to generating large numbers of megakaryocytes in culture. The genes responsible have been rigorously studied in vivo in mice, but are poorly characterized in human culture systems. As CD34-positive (
+ ) cells isolated from human umbilical vein cord blood were differentiated into megakaryocytes in culture, two distinct cell populations were identified by flow cytometric forward and side scatter: larger size, lower granularity (LLG), and smaller size, higher granularity (SHG). The LLG cells were CD41aHigh CD42aHigh phosphatidylserineLow , had an electron microscopic morphology similar to mature bone marrow megakaryocytes, developed proplatelets, and displayed a signaling response to platelet agonists. The SHG cells were CD41aLow CD42aLow phosphatidylserineHigh , had a distinctly apoptotic morphology, were unable to develop proplatelets, and showed no signaling response. Screens of differentiating megakaryocytes for expression of 24 apoptosis genes identified BCL2L2 as a novel candidate megakaryocyte apoptosis regulator. Lentiviral BCL2L2 overexpression decreased megakaryocyte apoptosis, increased CD41a+ LLG cells, and increased proplatelet formation by 58%. An association study in 154 healthy donors identified a significant positive correlation between platelet number and platelet BCL2L2 mRNA levels. This finding was consistent with the observed increase in platelet-like particles derived from cultured megakaryocytes over-expressing BCL2L2 BCL2L2 also induced small, but significant increases in thrombin-induced platelet-like particle αIIbβ3 activation and P-selectin expression. Thus, BCL2L2 restrains apoptosis in cultured megakaryocytes, promotes proplatelet formation, and is associated with platelet number. BCL2L2 is a novel target for improving megakaryocyte and platelet yields in in vitro culture systems., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
36. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets.
- Author
-
Cunin P, Bouslama R, Machlus KR, Martínez-Bonet M, Lee PY, Wactor A, Nelson-Maney N, Morris A, Guo L, Weyrich A, Sola-Visner M, Boilard E, Italiano JE, and Nigrovic PA
- Subjects
- Animals, Bone Marrow Cells metabolism, CD18 Antigens genetics, Cell Communication, Cytoplasm genetics, Cytoplasm metabolism, Cytoskeletal Proteins genetics, Humans, Inflammation blood, Inflammation pathology, Intercellular Adhesion Molecule-1 genetics, Membrane Transport Proteins genetics, Mice, Neutrophils metabolism, Blood Platelets metabolism, Emperipolesis genetics, Inflammation genetics, Megakaryocytes metabolism
- 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., Competing Interests: PC, RB, KM, MM, PL, AW, NN, AM, LG, AW, MS, EB, PN No competing interests declared, JI Is a founder of and has financial interest in Platelet BioGenesis, a company that aims to produce donor-independent human platelets from human induced pluripotent stem cells at scale. JEI's interests were reviewed and are managed by the Brigham and Women's Hospital and Partners HealthCare, in accordance with their conflict-of-interest policies., (© 2019, Cunin et al.)
- Published
- 2019
- Full Text
- View/download PDF
37. Aspirin inhibits platelets from reprogramming breast tumor cells and promoting metastasis.
- Author
-
Johnson KE, Ceglowski JR, Roweth HG, Forward JA, Tippy MD, El-Husayni S, Kulenthirarajan R, Malloy MW, Machlus KR, Chen WY, Italiano JE Jr, and Battinelli EM
- Subjects
- Blood Platelets metabolism, Breast Neoplasms metabolism, Cell Line, Tumor, Cytokines metabolism, Female, Humans, Neoplasm Metastasis, Neoplasm Staging, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-akt pharmacology, Signal Transduction, Aspirin pharmacology, Blood Platelets drug effects, Breast Neoplasms blood, Breast Neoplasms pathology, Platelet Aggregation Inhibitors pharmacology
- 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., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
38. Unlocking the Molecular Secrete(s) of α-Granule Biogenesis.
- Author
-
Noetzli LJ and Italiano JE Jr
- Subjects
- von Willebrand Factor, Blood Proteins, Megakaryocytes
- Published
- 2018
- Full Text
- View/download PDF
39. Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration.
- Author
-
Cloutier N, Allaeys I, Marcoux G, Machlus KR, Mailhot B, Zufferey A, Levesque T, Becker Y, Tessandier N, Melki I, Zhi H, Poirier G, Rondina MT, Italiano JE, Flamand L, McKenzie SE, Cote F, Nieswandt B, Khan WI, Flick MJ, Newman PJ, Lacroix S, Fortin PR, and Boilard E
- Subjects
- Adult, Anaphylaxis blood, Anaphylaxis genetics, Animals, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Platelet Activation, Platelet Count, Platelet Glycoprotein GPIIb-IIIa Complex genetics, Platelet Glycoprotein GPIIb-IIIa Complex immunology, Receptors, IgG genetics, Receptors, IgG immunology, Shock, Septic blood, Shock, Septic genetics, Young Adult, Anaphylaxis immunology, Antigen-Antibody Complex immunology, Blood Platelets immunology, Serotonin immunology, Shock, Septic immunology
- 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., Competing Interests: Conflict of interest statement: J.E.I. has a financial interest in and is a founder of Platelet BioGenesis, a company that aims to produce donor-independent human platelets from human-induced pluripotent stem cells at scale. J.E.I. is an inventor on this patent. The interests of J.E.I. were reviewed and are managed by the Brigham and Women’s Hospital and Partners HealthCare in accordance with their conflict of interest policies.
- Published
- 2018
- Full Text
- View/download PDF
40. Mature murine megakaryocytes present antigen-MHC class I molecules to T cells and transfer them to platelets.
- Author
-
Zufferey A, Speck ER, Machlus KR, Aslam R, Guo L, McVey MJ, Kim M, Kapur R, Boilard E, Italiano JE Jr, and Semple JW
- 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., Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.- Published
- 2017
- Full Text
- View/download PDF
41. Selinexor-induced thrombocytopenia results from inhibition of thrombopoietin signaling in early megakaryopoiesis.
- Author
-
Machlus KR, Wu SK, Vijey P, Soussou TS, Liu ZJ, Shacham E, Unger TJ, Kashyap T, Klebanov B, Sola-Visner M, Crochiere M, Italiano JE Jr, and Landesman Y
- Subjects
- Animals, Apoptosis drug effects, Blood Platelets drug effects, Blood Platelets pathology, Bone Marrow drug effects, Bone Marrow pathology, Cell Count, Cell Differentiation drug effects, Dose-Response Relationship, Drug, Fetus pathology, Liver embryology, Megakaryocytes drug effects, Megakaryocytes ultrastructure, Mice, Knockout, Platelet Activation drug effects, Stem Cells cytology, Thrombocytopenia blood, Hydrazines adverse effects, Megakaryocytes metabolism, Megakaryocytes pathology, Signal Transduction drug effects, Thrombocytopenia chemically induced, Thrombocytopenia metabolism, Thrombopoiesis drug effects, Thrombopoietin metabolism, Triazoles adverse effects
- Abstract
Selinexor is the first oral selective inhibitor of nuclear export compound tested for cancer treatment. Selinexor has demonstrated a safety therapy profile with broad antitumor activity against solid and hematological malignancies in phases 2 and 3 clinical trials (#NCT03071276, #NCT02343042, #NCT02227251, #NCT03110562, and #NCT02606461). Although selinexor shows promising efficacy, its primary adverse effect is high-grade thrombocytopenia. Therefore, we aimed to identify the mechanism of selinexor-induced thrombocytopenia to relieve it and improve its clinical management. We determined that selinexor causes thrombocytopenia by blocking thrombopoietin (TPO) signaling and therefore differentiation of stem cells into megakaryocytes. We then used both in vitro and in vivo models and patient samples to show that selinexor-induced thrombocytopenia is indeed reversible when TPO agonists are administered in the absence of selinexor (drug holiday). In sum, these data reveal (1) the mechanism of selinexor-induced thrombocytopenia, (2) an effective way to reverse the dose-limiting thrombocytopenia, and (3) a novel role for XPO1 in megakaryopoiesis. The improved selinexor dosing regimen described herein is crucial to help reduce thrombocytopenia in selinexor patients, allowing them to continue their course of chemotherapy and have the best chance of survival. This trial was registered at www.clinicaltrials.gov as #NCT01607905., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
42. Deletion of the Arp2/3 complex in megakaryocytes leads to microthrombocytopenia in mice.
- Author
-
Paul DS, Casari C, Wu C, Piatt R, Pasala S, Campbell RA, Poe KO, Ghalloussi D, Lee RH, Rotty JD, Cooley BC, Machlus KR, Italiano JE Jr, Weyrich AS, Bear JE, and Bergmeier W
- 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. Arpc2fl/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 Arpc2fl/fl PF4-Cre mice. Arpc2fl/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 Arpc2fl/fl PF4-Cre and control mice. Thus, Arp2/3 is critical for platelet homeostasis but plays only a minor role for vascular hemostasis., Competing Interests: Disclosure of Conflict of Interest J.E.I. has financial interest in and is a founder of Platelet BioGenesis, a company that aims to produce donor-independent human platelets from human-induced pluripotent stem cells at scale. J.E.I. is an inventor on this patent. The interests of J.E.I. were reviewed and are managed by the Brigham and Women’s Hospital and Partners HealthCare in accordance with their conflict-of-interest policies. The remaining authors have declared that no conflict of interest exists.- Published
- 2017
- Full Text
- View/download PDF
43. Megakaryocytes compensate for Kit insufficiency in murine arthritis.
- Author
-
Cunin P, Penke LR, Thon JN, Monach PA, Jones T, Chang MH, Chen MM, Melki I, Lacroix S, Iwakura Y, Ware J, Gurish MF, Italiano JE, Boilard E, and Nigrovic PA
- Subjects
- Animals, Fibroblasts immunology, Fibroblasts pathology, Immunoglobulin G immunology, Interleukin-1 genetics, Interleukin-1 immunology, Mast Cells immunology, Mast Cells pathology, Mice, Mice, Knockout, NF-E2 Transcription Factor, p45 Subunit genetics, NF-E2 Transcription Factor, p45 Subunit immunology, Platelet Membrane Glycoproteins genetics, Platelet Membrane Glycoproteins immunology, Arthritis, Experimental genetics, Arthritis, Experimental immunology, Arthritis, Experimental pathology, Megakaryocytes immunology, Megakaryocytes pathology, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit immunology, Synovial Membrane immunology, Synovial Membrane pathology
- Abstract
The growth factor receptor Kit is involved in hematopoietic and nonhematopoietic development. Mice bearing Kit defects lack mast cells; however, strains bearing different Kit alleles exhibit diverse phenotypes. Herein, we investigated factors underlying differential sensitivity to IgG-mediated arthritis in 2 mast cell-deficient murine lines: KitWsh/Wsh, which develops robust arthritis, and KitW/Wv, which does not. Reciprocal bone marrow transplantation between KitW/Wv and KitWsh/Wsh mice revealed that arthritis resistance reflects a hematopoietic defect in addition to mast cell deficiency. In KitW/Wv mice, restoration of susceptibility to IgG-mediated arthritis was neutrophil independent but required IL-1 and the platelet/megakaryocyte markers NF-E2 and glycoprotein VI. In KitW/Wv mice, platelets were present in numbers similar to those in WT animals and functionally intact, and transfer of WT platelets did not restore arthritis susceptibility. These data implicated a platelet-independent role for the megakaryocyte, a Kit-dependent lineage that is selectively deficient in KitW/Wv mice. Megakaryocytes secreted IL-1 directly and as a component of circulating microparticles, which activated synovial fibroblasts in an IL-1-dependent manner. Transfer of WT but not IL-1-deficient megakaryocytes restored arthritis susceptibility to KitW/Wv mice. These findings identify functional redundancy among Kit-dependent hematopoietic lineages and establish an unanticipated capacity of megakaryocytes to mediate IL-1-driven systemic inflammatory disease.
- Published
- 2017
- Full Text
- View/download PDF
44. Tamoxifen Directly Inhibits Platelet Angiogenic Potential and Platelet-Mediated Metastasis.
- Author
-
Johnson KE, Forward JA, Tippy MD, Ceglowski JR, El-Husayni S, Kulenthirarajan R, Machlus KR, Mayer EL, Italiano JE Jr, and Battinelli EM
- Subjects
- Blood Platelets metabolism, Breast Neoplasms blood, Breast Neoplasms pathology, Cell Proliferation drug effects, Coculture Techniques, Female, Human Umbilical Vein Endothelial Cells metabolism, Humans, MCF-7 Cells, Neoplasm Metastasis, Signal Transduction drug effects, Tamoxifen pharmacology, Vascular Endothelial Growth Factor A metabolism, Angiogenesis Inhibitors pharmacology, Blood Platelets drug effects, Breast Neoplasms drug therapy, Cell Movement drug effects, Human Umbilical Vein Endothelial Cells drug effects, Neovascularization, Physiologic drug effects, Platelet Activation drug effects, Platelet Aggregation Inhibitors pharmacology, Tamoxifen analogs & derivatives
- Abstract
Objective: Platelets, which are mainly known for their role in hemostasis, are now known to play a crucial role in metastasis. Tamoxifen is a selective estrogen receptor modulator that is widely used for the treatment of breast cancer. Tamoxifen and its metabolites have been shown to directly impact platelet function, suggesting that this drug has additional mechanisms of action. The purpose of this study was to determine whether tamoxifen exerts antitumor effects through direct platelet inhibition., Approach and Results: This study found that pretreatment with tamoxifen leads to a significant inhibition of platelet activation. Platelets exposed to tamoxifen released significantly lower amounts of proangiogenic regulator vascular endothelial growth factor. In vitro angiogenesis assays confirmed that tamoxifen pretreatment led to diminished capillary tube formation and decreased endothelial migration. Tamoxifen and its metabolite, 4-hydroxytamoxifen, also significantly inhibited the ability of platelets to promote metastasis in vitro. Using a membrane-based array, we identified several proteins associated with angiogenesis metastasis that were lower in activated releasate from tamoxifen-treated platelets, including angiogenin, chemokine (C-X-C motif) ligand 1, chemokine (C-C motif) ligand 5, epidermal growth factor, chemokine (C-X-C motif) ligand 5, platelet-derived growth factor dimeric isoform BB, whereas antiangiogenic angiopoietin-1 was elevated. Platelets isolated from patients on tamoxifen maintenance therapy were also found to have decreased activation responses, diminished vascular endothelial growth factor release, and lower angiogenic and metastatic potential., Conclusions: We demonstrate that tamoxifen and its metabolite 4-hydroxytamoxifen directly alter platelet function leading to decreased angiogenic and metastatic potential. Furthermore, this study supports the idea of utilizing targeted platelet therapies to inhibit the platelet's role in angiogenesis and malignancy., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
45. DREAM plays an important role in platelet activation and thrombogenesis.
- Author
-
Kim K, Tseng A, Barazia A, Italiano JE, and Cho J
- Subjects
- Animals, Blood Platelets metabolism, Disease Models, Animal, Flow Cytometry, Humans, Immunoblotting, Immunoprecipitation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Polymerase Chain Reaction, Signal Transduction physiology, Kv Channel-Interacting Proteins metabolism, Platelet Activation physiology, Repressor Proteins metabolism, Thrombosis metabolism
- Abstract
Downstream regulatory element antagonist modulator (DREAM), a transcriptional repressor, is known to modulate pain responses. However, it is unknown whether DREAM is expressed in anucleate platelets and plays a role in thrombogenesis. By using intravital microscopy with DREAM-null mice and their bone marrow chimeras, we demonstrated that both hematopoietic and nonhematopoietic cell DREAMs are required for platelet thrombus formation following laser-induced arteriolar injury. In a FeCl
3 -induced thrombosis model, we found that compared with wild-type (WT) control and nonhematopoietic DREAM knockout (KO) mice, DREAM KO control and hematopoietic DREAM KO mice showed a significant delay in time to occlusion. Tail bleeding time was prolonged in DREAM KO control mice, but not in WT or DREAM bone marrow chimeric mice. In vivo adoptive transfer experiments further indicated the importance of platelet DREAM in thrombogenesis. We found that DREAM deletion does not alter the ultrastructural features of platelets but significantly impairs platelet aggregation and adenosine triphosphate secretion induced by numerous agonists (collagen-related peptide, adenosine 5'-diphosphate, A23187, thrombin, or U46619). Biochemical studies revealed that platelet DREAM positively regulates phosphoinositide 3-kinase (PI3K) activity during platelet activation. Using DREAM-null platelets and PI3K isoform-specific inhibitors, we observed that platelet DREAM is important for α-granule secretion, Ca2+ mobilization, and aggregation through PI3K class Iβ (PI3K-Iβ). Genetic and pharmacological studies in human megakaryoblastic MEG-01 cells showed that DREAM is important for A23187-induced Ca2+ mobilization and its regulatory function requires Ca2+ binding and PI3K-Iβ activation. These results suggest that platelet DREAM regulates PI3K-Iβ activity and plays an important role during thrombus formation., (© 2017 by The American Society of Hematology.)- Published
- 2017
- Full Text
- View/download PDF
46. Human thrombopoiesis depends on Protein kinase Cδ/protein kinase Cε functional couple.
- Author
-
Carubbi C, Masselli E, Martini S, Galli D, Aversa F, Mirandola P, Italiano JE Jr, Gobbi G, and Vitale M
- Subjects
- Adult, Aged, Blood Platelets metabolism, Cell Differentiation genetics, Female, Gene Expression, Gene Expression Regulation, Humans, Male, Megakaryocytes cytology, Megakaryocytes metabolism, Middle Aged, Primary Myelofibrosis blood, Primary Myelofibrosis diagnosis, Primary Myelofibrosis metabolism, Protein Binding, Protein Kinase C-delta genetics, Protein Kinase C-epsilon genetics, Thrombocythemia, Essential blood, Thrombocythemia, Essential diagnosis, Thrombocythemia, Essential metabolism, bcl-2-Associated X Protein genetics, bcl-2-Associated X Protein metabolism, bcl-X Protein genetics, bcl-X Protein metabolism, Protein Kinase C-delta metabolism, Protein Kinase C-epsilon metabolism, Thrombopoiesis genetics
- Abstract
A deeper understanding of the molecular events driving megakaryocytopoiesis and thrombopoiesis is essential to regulate in vitro and in vivo platelet production for clinical applications. We previously documented the crucial role of PKCε in the regulation of human and mouse megakaryocyte maturation and platelet release. However, since several data show that different PKC isoforms fulfill complementary functions, we targeted PKCε and PKCδ, which show functional and phenotypical reciprocity, at the same time as boosting platelet production in vitro. Results show that PKCδ, contrary to PKCε, is persistently expressed during megakaryocytic differentiation, and a forced PKCδ down-modulation impairs megakaryocyte maturation and platelet production. PKCδ and PKCε work as a functional couple with opposite roles on thrombopoiesis, and the modulation of their balance strongly impacts platelet production. Indeed, we show an imbalance of PKCδ/PKCε ratio both in primary myelofibrosis and essential thrombocythemia, featured by impaired megakaryocyte differentiation and increased platelet production, respectively. Finally, we demonstrate that concurrent molecular targeting of both PKCδ and PKCε represents a strategy for in vitro platelet factories., (Copyright© Ferrata Storti Foundation.)
- Published
- 2016
- Full Text
- View/download PDF
47. Lysyl oxidase is associated with increased thrombosis and platelet reactivity.
- Author
-
Matsuura S, Mi R, Koupenova M, Eliades A, Patterson S, Toselli P, Thon J, Italiano JE Jr, Trackman PC, Papadantonakis N, and Ravid K
- Subjects
- Animals, Blood Platelets cytology, Carotid Artery Injuries complications, Carotid Artery Thrombosis etiology, Integrin alpha2beta1 physiology, Megakaryocytes enzymology, Mice, Mice, Transgenic, Peptide Fragments pharmacology, Platelet Adhesiveness genetics, Platelet Adhesiveness physiology, Platelet Aggregation drug effects, Platelet Aggregation genetics, Platelet Factor 4 genetics, Promoter Regions, Genetic, Protein-Lysine 6-Oxidase genetics, Rats, Thrombophilia genetics, Blood Platelets drug effects, Collagen pharmacology, Platelet Activation physiology, Protein-Lysine 6-Oxidase physiology, Thrombophilia enzymology
- 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., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
48. Synthesis and dephosphorylation of MARCKS in the late stages of megakaryocyte maturation drive proplatelet formation.
- Author
-
Machlus KR, Wu SK, Stumpo DJ, Soussou TS, Paul DS, Campbell RA, Kalwa H, Michel T, Bergmeier W, Weyrich AS, Blackshear PJ, Hartwig JH, and Italiano JE Jr
- Subjects
- Actin-Related Protein 2-3 Complex metabolism, Actin-Related Protein 3 metabolism, Amino Acid Sequence, Angiopoietin-Like Protein 2, Angiopoietin-like Proteins, Angiopoietins metabolism, Animals, Apoptosis, Blood Platelets metabolism, Intracellular Signaling Peptides and Proteins deficiency, Liver cytology, Liver embryology, Membrane Proteins deficiency, Membrane Proteins metabolism, Mice, Mice, Knockout, Molecular Sequence Data, Myristoylated Alanine-Rich C Kinase Substrate, Peptide Fragments metabolism, Peptide Fragments pharmacology, Phosphatidylinositol 4,5-Diphosphate metabolism, Phosphorylation, Protein Biosynthesis, Protein Kinase C metabolism, Signal Transduction, Intracellular Signaling Peptides and Proteins physiology, Megakaryocytes metabolism, Membrane Proteins physiology, Protein Processing, Post-Translational, Thrombopoiesis physiology
- Abstract
Platelets are essential for hemostasis, and thrombocytopenia is a major clinical problem. Megakaryocytes (MKs) generate platelets by extending long processes, proplatelets, into sinusoidal blood vessels. However, very little is known about what regulates proplatelet formation. To uncover which proteins were dynamically changing during this process, we compared the proteome and transcriptome of round vs proplatelet-producing MKs by 2D difference gel electrophoresis (DIGE) and polysome profiling, respectively. Our data revealed a significant increase in a poorly-characterized MK protein, myristoylated alanine-rich C-kinase substrate (MARCKS), which was upregulated 3.4- and 5.7-fold in proplatelet-producing MKs in 2D DIGE and polysome profiling analyses, respectively. MARCKS is a protein kinase C (PKC) substrate that binds PIP2. In MKs, it localized to both the plasma and demarcation membranes. MARCKS inhibition by peptide significantly decreased proplatelet formation 53%. To examine the role of MARCKS in the PKC pathway, we treated MKs with polymethacrylate (PMA), which markedly increased MARCKS phosphorylation while significantly inhibiting proplatelet formation 84%, suggesting that MARCKS phosphorylation reduces proplatelet formation. We hypothesized that MARCKS phosphorylation promotes Arp2/3 phosphorylation, which subsequently downregulates proplatelet formation; both MARCKS and Arp2 were dephosphorylated in MKs making proplatelets, and Arp2 inhibition enhanced proplatelet formation. Finally, we used MARCKS knockout (KO) mice to probe the direct role of MARCKS in proplatelet formation; MARCKS KO MKs displayed significantly decreased proplatelet levels. MARCKS expression and signaling in primary MKs is a novel finding. We propose that MARCKS acts as a "molecular switch," binding to and regulating PIP2 signaling to regulate processes like proplatelet extension (microtubule-driven) vs proplatelet branching (Arp2/3 and actin polymerization-driven).
- Published
- 2016
- Full Text
- View/download PDF
49. CCL5 derived from platelets increases megakaryocyte proplatelet formation.
- Author
-
Machlus KR, Johnson KE, Kulenthirarajan R, Forward JA, Tippy MD, Soussou TS, El-Husayni SH, Wu SK, Wang S, Watnick RS, Italiano JE Jr, and Battinelli EM
- Subjects
- Animals, Blood Platelets cytology, Chemokine CCL5 genetics, Cyclohexanes pharmacology, Humans, Maraviroc, Megakaryocytes cytology, Mice, Receptors, CCR5 genetics, Receptors, CCR5 metabolism, Signal Transduction drug effects, Triazoles pharmacology, Blood Platelets metabolism, Chemokine CCL5 metabolism, Megakaryocytes pathology, Signal Transduction physiology
- Abstract
In times of physiological stress, platelet count can transiently rise. What initiates this reactive thrombocytosis is poorly understood. Intriguingly, we found that treating megakaryocytes (MKs) with the releasate from activated platelets increased proplatelet production by 47%. Platelets store inflammatory cytokines, including the chemokine ligand 5 (CCL5, RANTES); after TRAP activation, platelets release over 25 ng/mL CCL5. We hypothesized that CCL5 could regulate platelet production by binding to its receptor, CCR5, on MKs. Maraviroc (CCR5 antagonist) or CCL5 immunodepletion diminished 95% and 70% of the effect of platelet releasate, respectively, suggesting CCL5 derived from platelets is sufficient to drive increased platelet production through MK CCR5. MKs cultured with recombinant CCL5 increased proplatelet production by 50% and had significantly higher ploidy. Pretreating the MK cultures with maraviroc prior to exposure to CCL5 reversed the augmented proplatelet formation and ploidy, suggesting that CCL5 increases MK ploidy and proplatelet formation in a CCR5-dependent manner. Interrogation of the Akt signaling pathway suggested that CCL5/CCR5 may influence proplatelet production by suppressing apoptosis. In an in vivo murine acute colitis model, platelet count significantly correlated with inflammation whereas maraviroc treatment abolished this correlation. We propose that CCL5 signaling through CCR5 may increase platelet counts during physiological stress., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
50. Cytoskeletal perturbation leads to platelet dysfunction and thrombocytopenia in variant forms of Glanzmann thrombasthenia.
- Author
-
Bury L, Falcinelli E, Chiasserini D, Springer TA, Italiano JE Jr, and Gresele P
- Subjects
- Animals, Blood Platelets pathology, CHO Cells, Cricetinae, Cricetulus, Cytoskeleton genetics, Cytoskeleton pathology, Female, Humans, Integrin alpha2 genetics, Integrin alpha2 metabolism, Integrin beta3 genetics, Integrin beta3 metabolism, Male, Thrombasthenia genetics, Thrombasthenia pathology, Thrombocytopenia genetics, Thrombocytopenia pathology, Blood Platelets metabolism, Cytoskeleton metabolism, Mutation, Thrombasthenia metabolism, Thrombocytopenia metabolism
- Abstract
Several patients have been reported to have variant dominant forms of Glanzmann thrombasthenia, associated with macrothrombocytopenia and caused by gain-of-function mutations of ITGB3 or ITGA2B leading to reduced surface expression and constitutive activation of integrin αIIbβ3. The mechanisms leading to a bleeding phenotype of these patients have never been addressed. The aim of this study was to unravel the mechanism by which ITGB3 mutations causing activation of αIIbβ3 lead to platelet dysfunction and macrothrombocytopenia. Using platelets from two patients carrying the β3 del647-686 mutation and Chinese hamster ovary cells expressing different αIIbβ3-activating mutations, we showed that reduced surface expression of αIIbβ3 is due to receptor internalization. Moreover, we demonstrated that permanent triggering of αIIbβ3-mediated outside-in signaling causes an impairment of cytoskeletal reorganization arresting actin turnover at the stage of polymerization. The induction of actin polymerization by jasplakinolide, a natural toxin that promotes actin nucleation and prevents depolymerization of stress fibers, in control platelets produced an impairment of platelet function similar to that of patients with variant forms of dominant Glanzmann thrombasthenia. del647-686β3-transduced murine megakaryocytes generated proplatelets with a reduced number of large tips and asymmetric barbell-proplatelets, suggesting that impaired cytoskeletal rearrangement is the cause of macrothrombocytopenia. These data show that impaired cytoskeletal remodeling caused by a constitutively activated αIIbβ3 is the main effector of platelet dysfunction and macrothrombocytopenia, and thus of bleeding, in variant forms of dominant Glanzmann thrombasthenia., (Copyright© Ferrata Storti Foundation.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.