Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
Biochem Biophys Res Commun ; 585: 96-102, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34801938

ABSTRACT

Platelet plays an important role in the progression of atherosclerosis. Recently it has been reported that myocardial infarction (MI) triggers megakaryopoiesis and thrombopoiesis in the bone marrow and leads to increased circulating platelets, which might contribute to the aggravation of atherosclerosis. However, the underlying mechanisms remain unclear. Here, we analyzed post-MI bone marrow tissue and found that MI induced an upregulation of bone marrow NOD-like Receptor Protein 3 (NLRP3) and subsequent secretion of IL-1ß, an essential stimulator of megakaryopoiesis. Targeting NLRP3 using a specific inhibitor MCC950 reduced bone marrow IL-1ß expression. Using bone marrow whole-mount immunofluorescence staining combined with flow cytometry, we demonstrated that MCC950 reduced megakaryocyte cellularity and maturity, and effectively attenuated the excessive platelet production after MI. Importantly, mice subjected to MI treated with MCC950 showed a higher survival rate compared with the only MI group. Taken together, this study shows that bone marrow NLRP3-IL-1ß signal regulates megakaryocyte development and platelet production after myocardial infarction. It provides a new hint that pharmacological inhibition of NLRP3 might become a potential therapeutic approach for controlling excessive thrombopoiesis after MI.


Subject(s)
Bone Marrow/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Megakaryocytes/metabolism , Myocardial Infarction/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Thrombopoiesis/physiology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Flow Cytometry , Furans/pharmacology , Indenes/pharmacology , Inflammasomes/drug effects , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Survival Analysis , Thrombopoiesis/drug effects
2.
Arterioscler Thromb Vasc Biol ; 41(11): 2661-2670, 2021 11.
Article in English | MEDLINE | ID: mdl-34615371

ABSTRACT

Myeloproliferative neoplasms (MPNs) are a group of malignant disorders of the bone marrow where a dysregulated balance between proliferation and differentiation gives rise to abnormal numbers of mature blood cells. MPNs encompass a spectrum of disease entities with progressively more severe clinical features, including complications with thrombosis and hemostasis and an increased propensity for transformation to acute myeloid leukemia. There is an unmet clinical need for markers of disease progression. Our understanding of the precise mechanisms that influence pathogenesis and disease progression has been limited by access to disease-specific cells as biosources. Here, we review the landscape of MPN pathology and present blood platelets as potential candidates for disease-specific understanding. We conclude with our recent work discovering progressive platelet heterogeneity by subtype in a large clinical cohort of patients with MPN.


Subject(s)
Blood Platelets/metabolism , Myeloproliferative Disorders/blood , Platelet Activation , Thrombopoiesis , Animals , Antineoplastic Agents/therapeutic use , Blood Coagulation , Blood Platelets/drug effects , Blood Platelets/pathology , Humans , Molecular Targeted Therapy , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phenotype , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Thrombopoiesis/drug effects
3.
Sci Rep ; 11(1): 17990, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504210

ABSTRACT

Macrothrombocytopenia is a common pathology of missense mutations in genes regulating actin dynamics. Takenouchi-Kosaki syndrome (TKS) harboring the c.191A > G, Tyr64Cys (Y64C) variant in Cdc42 exhibits a variety of clinical manifestations, including immunological and hematological anomalies. In the present study, we investigated the functional abnormalities of the Y64C mutant in HEK293 cells and elucidated the mechanism of macrothrombocytopenia, one of the symptoms of TKS patients, by monitoring the production of platelet-like particles (PLP) using MEG-01 cells. We found that the Y64C mutant was concentrated at the membrane compartment due to impaired binding to Rho-GDI and more active than the wild-type. The Y64C mutant also had lower association with its effectors Pak1/2 and N-WASP. Y64C mutant-expressing MEG-01 cells demonstrated short cytoplasmic protrusions with aberrant F-actin and microtubules, and reduced PLP production. This suggested that the Y64C mutant facilitates its activity and membrane localization, resulting in impaired F-actin dynamics for proplatelet extension, which is necessary for platelet production. Furthermore, such dysfunction was ameliorated by either suppression of Cdc42 activity or prenylation using chemical inhibitors. Our study may lead to pharmacological treatments for TKS patients.


Subject(s)
Megakaryocytes/drug effects , Megakaryocytes/metabolism , Signal Transduction/drug effects , Thrombocytopenia/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Benzamides/pharmacology , Blood Platelets/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Membrane/metabolism , HEK293 Cells , Humans , Mutation , Protein Prenylation/drug effects , Pyrazoles/pharmacology , Signal Transduction/genetics , Sulfonamides/pharmacology , Syndrome , Thrombocytopenia/genetics , Thrombopoiesis/drug effects , Thrombopoiesis/genetics , Transfection , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
4.
Arterioscler Thromb Vasc Biol ; 41(10): 2523-2537, 2021 10.
Article in English | MEDLINE | ID: mdl-34348487

ABSTRACT

Objective: Roxadustat is a new medication for the treatment of renal anemia. EPO (erythropoietin)-the current treatment standard-has been reported to enhance platelet activation and production. However, to date, the effect of roxadustat on platelets is unclear. To address this deficiency, herein, we have evaluated the effect of roxadustat on platelet production and function. Approach and Results: We performed several mouse platelet functional assays in the presence/absence of in vitro and in vivo roxadustat treatment. Both healthy and 5/6 nephrectomized mice were utilized. The effect of roxadustat on platelet function of healthy volunteers and chronic kidney disease patients was also evaluated. For platelet production, megakaryocyte maturation and proplatelet formation were assayed in vitro. Peripheral platelet and bone marrow megakaryocyte counts were also determined. We found that roxadustat could not stimulate washed platelets directly, and platelet aggregation, spreading, clot retraction, and P-selectin/JON/A exposure were similar with or without in vitro or in vivo roxadustat treatment among both healthy and 5/6 nephrectomized mice. In vivo mouse thrombosis models were additionally performed, and no differences were detected between the vehicle and roxadustat treatment groups. EPO, which was considered a positive control in the present study, promoted platelet function and production as reported previously. Megakaryocyte maturation and proplatelet formation were also not significantly different between control mice and those treated with roxadustat. After receiving roxadustat for 14 days, no difference in the peripheral platelet count was observed in the mice. Conclusions: Administration of roxadustat has no significant impact on platelet production and function.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/drug effects , Erythropoietin/pharmacology , Glycine/analogs & derivatives , Hematinics/pharmacology , Isoquinolines/pharmacology , Platelet Activation/drug effects , Thrombopoiesis/drug effects , Thrombosis/blood , Animals , Blood Platelets/metabolism , Case-Control Studies , Disease Models, Animal , Erythropoietin/toxicity , Glycine/pharmacology , Glycine/toxicity , Hematinics/toxicity , Humans , Isoquinolines/toxicity , Male , Mice, Inbred C57BL , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/etiology , Thrombosis/etiology
5.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34153091

ABSTRACT

Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E2 (dmPGE2), a longer-acting analogue of PGE2, promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE2 and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE2 and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE2 and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE2, alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin-CD45-CD31+Sca-1- sinusoidal endothelial cells, while combined dmPGE2 and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE2 and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.


Subject(s)
16,16-Dimethylprostaglandin E2/therapeutic use , Acute Radiation Syndrome/drug therapy , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Blood Platelets/drug effects , Endothelial Cells/drug effects , Hemorrhagic Disorders/drug therapy , Lisinopril/therapeutic use , Megakaryocytes/drug effects , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Acute Radiation Syndrome/complications , Animals , Blood Platelets/radiation effects , Bone Marrow/drug effects , Bone Marrow/radiation effects , C-Reactive Protein/analysis , Cesium Radioisotopes , Drug Evaluation, Preclinical , Endothelial Cells/radiation effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Female , Gamma Rays/adverse effects , Hemorrhagic Disorders/etiology , Megakaryocytes/radiation effects , Mice , Mice, Inbred C57BL , P-Selectin/analysis , Platelet Aggregation/drug effects , Platelet Aggregation/radiation effects , Platelet Factor 4/analysis , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/etiology , Thrombocytopenia/etiology , Thrombopoiesis/radiation effects , Whole-Body Irradiation , von Willebrand Factor/analysis
6.
Elife ; 102021 06 01.
Article in English | MEDLINE | ID: mdl-34059198

ABSTRACT

Thrombocytopenic disorders have been treated with the Thrombopoietin-receptor agonist Eltrombopag. Patients with the same apparent form of thrombocytopenia may respond differently to the treatment. We describe a miniaturized bone marrow tissue model that provides a screening bioreactor for personalized, pre-treatment response prediction to Eltrombopag for individual patients. Using silk fibroin, a 3D bone marrow niche was developed that reproduces platelet biogenesis. Hematopoietic progenitors were isolated from a small amount of peripheral blood of patients with mutations in ANKRD26 and MYH9 genes, who had previously received Eltrombopag. The ex vivo response was strongly correlated with the in vivo platelet response. Induced Pluripotent Stem Cells (iPSCs) from one patient with mutated MYH9 differentiated into functional megakaryocytes that responded to Eltrombopag. Combining patient-derived cells and iPSCs with the 3D bone marrow model technology allows having a reproducible system for studying drug mechanisms and for individualized, pre-treatment selection of effective therapy in Inherited Thrombocytopenias.


Platelets are tiny cell fragments essential for blood to clot. They are created and released into the bloodstream by megakaryocytes, giant cells that live in the bone marrow. In certain genetic diseases, such as Inherited Thrombocytopenia, the bone marrow fails to produce enough platelets: this leaves patients extremely susceptible to bruising, bleeding, and poor clotting after an injury or surgery. Certain patients with Inherited Thrombocytopenia respond well to treatments designed to boost platelet production, but others do not. Why these differences exist could be investigated by designing new test systems that recreate the form and function of bone marrow in the laboratory. However, it is challenging to build the complex and poorly understood bone marrow environment outside of the body. Here, Di Buduo et al. have developed an artificial three-dimensional miniature organ bioreactor system that recreates the key features of bone marrow. In this system, megakaryocytes were grown from patient blood samples, and hooked up to a tissue scaffold made of silk. The cells were able to grow as if they were in their normal environment, and they could shed platelets into an artificial bloodstream. After treating megakaryocytes with drugs to stimulate platelet production, Di Buduo et al. found that the number of platelets recovered from the bioreactor could accurately predict which patients would respond to these drugs in the clinic. This new test system enables researchers to predict how a patient will respond to treatment, and to tailor therapy options to each individual. This technology could also be used to test new drugs for Inherited Thrombocytopenias and other blood-related diseases; if scaled-up, it could also, one day, generate large quantities of lab-grown blood cells for transfusion.


Subject(s)
Benzoates/pharmacology , Blood Platelets/drug effects , Hematopoietic Stem Cells/drug effects , Hydrazines/pharmacology , Induced Pluripotent Stem Cells/drug effects , Megakaryocytes/drug effects , Pyrazoles/pharmacology , Receptors, Thrombopoietin/agonists , Stem Cell Niche , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Adult , Aged , Bioreactors , Blood Platelets/metabolism , Cell Culture Techniques , Cells, Cultured , Female , Fibroins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Megakaryocytes/metabolism , Middle Aged , Miniaturization , Mutation , Myosin Heavy Chains/genetics , Receptors, Thrombopoietin/metabolism , Thrombocytopenia/blood , Thrombocytopenia/genetics , Young Adult
7.
CPT Pharmacometrics Syst Pharmacol ; 10(5): 489-499, 2021 05.
Article in English | MEDLINE | ID: mdl-33797208

ABSTRACT

Platelets are produced by hematopoietic stem cells via megakaryocytes in the bone marrow and play a critical role in hemostasis. The aim of this study was to develop a new platelet model based on the thrombopoiesis and platelet life-cycle by a quantitative systems pharmacology modeling approach, which could describe changes in platelet count profiles in platelet-related diseases and drug intervention. The proposed platelet model consists of 44 components. The model was applied to thrombopoiesis of a thrombopoietin receptor agonist, lusutrombopag. It could well describe the observed platelet count profiles after administration of lusutrombopag for both healthy subjects and patients with chronic liver disease and thrombocytopenia. This model should be useful for understanding the disease progression of platelet-related conditions, such as thrombocytopenia and for predicting platelet count profiles in various disease situations related to platelets and drug administration in drug development.


Subject(s)
Blood Platelets/drug effects , Cinnamates/pharmacology , Computer Simulation , End Stage Liver Disease/drug therapy , Receptors, Thrombopoietin/agonists , Thiazoles/pharmacology , Thrombopoiesis/drug effects , Cinnamates/therapeutic use , Humans , Thiazoles/therapeutic use
8.
Cell Death Dis ; 12(2): 208, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627636

ABSTRACT

Chronic myeloid leukemia (CML) is characterized by the accumulation of malignant and immature white blood cells which spread to the peripheral blood and other tissues/organs. Despite the fact that current tyrosine kinase inhibitors (TKIs) are capable of achieving the complete remission by reducing the tumor burden, severe adverse effects often occur in CML patients treated with TKIs. The differentiation therapy exhibits therapeutic potential to improve cure rates in leukemia, as evidenced by the striking success of all-trans-retinoic acid in acute promyelocytic leukemia treatment. However, there is still a lack of efficient differentiation therapy strategy in CML. Here we showed that MPL, which encodes the thrombopoietin receptor driving the development of hematopoietic stem/progenitor cells, decreased along with the progression of CML. We first elucidated that MPL signaling blockade impeded the megakaryocytic differentiation and contributed to the progression of CML. While allogeneic human umbilical cord-derived mesenchymal stem cells (UC-MSCs) treatment efficiently promoted megakaryocytic lineage differentiation of CML cells through restoring the MPL expression and activating MPL signaling. UC-MSCs in combination with eltrombopag, a non-peptide MPL agonist, further activated JAK/STAT and MAPK signaling pathways through MPL and exerted a synergetic effect on enhancing CML cell differentiation. The established combinational treatment not only markedly reduced the CML burden but also significantly eliminated CML cells in a xenograft CML model. We provided a new molecular insight of thrombopoietin (TPO) and MPL signaling in MSCs-mediated megakaryocytic differentiation of CML cells. Furthermore, a novel anti-CML treatment regimen that uses the combination of UC-MSCs and eltrombopag shows therapeutic potential to overcome the differentiation blockade in CML.


Subject(s)
Benzoates/pharmacology , Hydrazines/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Pyrazoles/pharmacology , Receptors, Thrombopoietin/agonists , Thrombopoiesis/drug effects , Animals , Cell Lineage , Coculture Techniques , Gene Expression Regulation, Leukemic , Humans , Janus Kinases/metabolism , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Receptors, Thrombopoietin/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Umbilical Cord/cytology , Xenograft Model Antitumor Assays
9.
Mini Rev Med Chem ; 21(13): 1638-1645, 2021.
Article in English | MEDLINE | ID: mdl-33390132

ABSTRACT

Pluripotent stem cells of the bone marrow are stimulated by different cytokines to proliferation and differentiation into various types of blood cells. These cytokines are mostly glycoproteins. Erythropoietin stimulates stem cells to the formation of erythrocytes while colony-stimulating factors cause the formation of different types of white blood cells. Stem cell factors play an important role in the maintenance and survival of blood cells of all types. Thrombopoietin stimulates stem cells to proliferation and formation of blood platelets. Granulocyte colony-stimulating factor is probably the most important drug in use. It stimulates stem cells to the formation of neutrophile granulocytes. It is often used in recombinant forms such as filgrastim in the treatment of neutropenia in cancer chemotherapy or AIDS. Its pegylated conjugates such as pegfilgrastim are also available. Its activity can be supported by plerixafor, a small molecule - bicyclam derivative acting as an indirect agonist of stem cells factor. It acts as an antagonist of CXCR4 receptor activation of which brakes hematopoiesis. The treatment of conditions accompanied by thrombocytopenia such as idiopathic thrombocytopenic purpura is currently not performed by thrombopoietin but synthetic agonists of its receptor are preferred. Romiplostim is a peptibody. It consists of a protein part interacting with the thrombopoietin receptor which is, however, different from thrombopoietin, and of Fc fragment of immunoglobulin G1. In contrast, small molecule thrombopoietin receptor agonists represented by eltrombopag can be given orally unlike all of the above.


Subject(s)
Colony-Stimulating Factors/pharmacology , Small Molecule Libraries/pharmacology , Stem Cell Factor/pharmacology , Thrombopoiesis/drug effects , Benzoates/chemistry , Benzoates/pharmacology , Blood Platelets/cytology , Blood Platelets/metabolism , Cell Differentiation/drug effects , Humans , Hydrazines/chemistry , Hydrazines/pharmacology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Receptors, Thrombopoietin/agonists , Receptors, Thrombopoietin/metabolism , Small Molecule Libraries/chemistry
10.
Thromb Haemost ; 121(2): 192-205, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32961571

ABSTRACT

Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase, has a favorable safety profile in patients with B cell-related malignancies. A primary adverse effect of ibrutinib is thrombocytopenia in the early stages of treatment, but platelet counts increase or recover as treatment continues. Currently, the effects of ibrutinib on megakaryopoiesis remain unclear. In this study, we investigated the mechanism by which ibrutinib induces thrombocytopenia using cord blood CD34+ hematopoietic stem cells (HSCs), a human megakaryoblastic cell line (SET-2), and C57BL/6 mice. We show that treatment with ibrutinib can suppress CD34+ HSC differentiation into megakaryocytes (MKs) and decrease the number of colony-forming unit-MKs (CFU-MKs). The ibrutinib-dependent inhibition of early megakaryopoiesis seems to mainly involve impaired proliferation of progenitor cells without induction of apoptosis. The effects of ibrutinib on late-stage megakaryopoiesis, in contrast to early-stage megakaryopoiesis, include enhanced MK differentiation, ploidy, and proplatelet formation in CD34+ HSC-derived MKs and SET-2 cells. We also demonstrated that MK adhesion and spreading, but not migration, were inhibited by ibrutinib. Furthermore, we revealed that integrin αIIbß3 outside-in signaling in MKs was inhibited by ibrutinib. Consistent with previous clinical observations, in C57BL/6 mice treated with ibrutinib, platelet counts decreased by days 2 to 7 and recovered to normal levels by day 15. Together, these results reveal the pathogenesis of ibrutinib-induced transient thrombocytopenia. In conclusion, ibrutinib suppresses early megakaryopoiesis, as evidenced by inhibition of MK progenitor cell proliferation and CFU-MK formation. Ibrutinib enhances MK differentiation, ploidy, and proplatelet formation, while it impairs integrin αIIbß3 outside-in signaling.


Subject(s)
Adenine/analogs & derivatives , Blood Platelets/drug effects , Megakaryocytes/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Thrombopoiesis/drug effects , Adenine/pharmacology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Blood Platelets/cytology , Cell Line , Humans , Megakaryocytes/cytology , Mice, Inbred C57BL
11.
Platelets ; 32(3): 378-382, 2021 Apr 03.
Article in English | MEDLINE | ID: mdl-32268817

ABSTRACT

Thrombocytopenia is a severe complication for patients with myelodysplastic syndrome (MDS). Eltrombopag increases platelet count in MDS patients but its combination with azacitidine elicited controversial results. We aimed to quantify the colony forming units of megakaryocytes (CFU-Mk) obtained from CD34+ bone marrow cells isolated from patients with MDS and from healthy donors that were cultured in vitro in the presence or absence of azacitidine and with or without the sequential addition of eltrombopag to the culture medium. CD34+ bone marrow cells from 6 MDS patients and 3 controls were expanded in vitro and cultured for 3 days with or without azacitidine. Subsequently, a CFU-Mk assay was performed in presence or absence of eltrombopag. The addition of eltrombopag in the CFU-Mk assay after mock treatment of CD34+ cells increased the number of CFU-Mk in both controls and patients. On the contrary, using azacitidine pretreated CD34+ cells, eltrombopag minimally increased CFU-Mk in controls and produced heterogeneous response in MDS patients with no change in two patients and CFU-Mk increase in four patients. In vitro CFU-Mk assay suggest that some MDS patients are likely to benefit from the sequential addition of eltrombopag after azacitidine treatment, in the context of a personalized medicine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/therapeutic use , Benzoates/therapeutic use , Hydrazines/therapeutic use , Myelodysplastic Syndromes/drug therapy , Pyrazoles/therapeutic use , Thrombopoiesis/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/pharmacology , Benzoates/pharmacology , Humans , Hydrazines/pharmacology , Middle Aged , Myelodysplastic Syndromes/pathology , Pyrazoles/pharmacology
12.
Biosci Rep ; 40(8)2020 08 28.
Article in English | MEDLINE | ID: mdl-32779719

ABSTRACT

Platelet transfusion is required for life-threatening thrombocytopenic bleeding, and single donor platelet concentrate is the ideal transfusion product. However, due to the inadequate number of donors that can donate a large volume of platelets, in vitro platelets production could be an alternative. We developed an in vitro production system designed to increase the platelet production yield from cultured cells. Previously, we reported that depletion of a Hippo pathway core kinase (LATS1/2) inhibited platelet production from cultured megakaryocytes. In the present study, we further investigated the role of the Hippo pathway in megakaryocyte proliferation and platelet production by focusing on the role of its effector proteins (YAP and TAZ), which are down-stream targets of LATS1/2 kinase. We found that YAP plays an essential role in megakaryoblastic cell proliferation, maturation, and platelet production, while TAZ showed minor effect. Knockdown of YAP, either by genetic manipulation or pharmaceutical molecule, significantly increased caspase-3-mediated apoptosis in cultured megakaryocytes, and increased platelet production as opposed to overexpressing YAP. We, therefore, demonstrate a paradigm for the regulation of megakaryocyte development and platelet production via the Hippo signaling pathway, and suggest the potential use of an FDA-approved drug to induce higher platelet production in cultured cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets/metabolism , Megakaryocytes/metabolism , Thrombopoiesis , Trans-Activators/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Blood Platelets/drug effects , Cell Line, Tumor , Dobutamine/pharmacology , Gene Expression Regulation , Humans , Megakaryocytes/drug effects , Signal Transduction , Thrombopoiesis/drug effects , Trans-Activators/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Verteporfin/pharmacology , YAP-Signaling Proteins
13.
Aging (Albany NY) ; 12(13): 13633-13646, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32651992

ABSTRACT

Clinical studies have shown that melatonin lowers the frequency of thrombocytopenia in patients with cancer undergoing radiotherapy or chemotherapy. Here, we investigated the mechanisms by which melatonin promotes platelet formation and survival. Our results show that melatonin exerted protective effects on serum-free induced apoptosis of CHRF megakaryocytes (MKs). Melatonin promoted the formation of MK colony forming units (CFUs) in a dose-dependent manner. Using doxorubicin-treated CHRF cells, we found that melatonin rescued G2/M cell cycle arrest and cell apoptosis induced by doxorubicin. The expression of p-AKT was increased by melatonin treatment, an effect that was abolished by melatonin receptor blocker. In addition, we demonstrated that melatonin enhanced the recovery of platelets in an irradiated mouse model. Megakaryopoiesis was largely preserved in melatonin-treated mice. We obtained the same results in vivo from bone marrow histology and CFU-MK formation assays. Melatonin may exert these protective effects by directly stimulating megakaryopoiesis and inhibiting megakaryocyte apoptosis through activation of its receptors and AKT signaling.


Subject(s)
Megakaryocytes/drug effects , Melatonin/pharmacology , Radiation Injuries, Experimental/prevention & control , Thrombocytopenia/prevention & control , Thrombopoiesis/drug effects , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Apoptosis/radiation effects , Blood Platelets/drug effects , Blood Platelets/physiology , Blood Platelets/radiation effects , Bone Marrow/drug effects , Bone Marrow/physiology , Bone Marrow/radiation effects , Caspases/metabolism , Cell Line, Tumor , Doxorubicin/adverse effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Megakaryocytes/physiology , Melatonin/therapeutic use , Mice , Mitochondria/metabolism , Neoplasms/therapy , Proto-Oncogene Proteins c-akt/metabolism , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/etiology , Receptors, Melatonin/antagonists & inhibitors , Receptors, Melatonin/metabolism , Stem Cells/drug effects , Thrombocytopenia/blood , Thrombocytopenia/etiology , Thrombopoiesis/radiation effects , Whole-Body Irradiation
14.
Clin Liver Dis ; 24(3): 437-451, 2020 08.
Article in English | MEDLINE | ID: mdl-32620282

ABSTRACT

Thrombocytopenia is common in advanced liver disease, and such patients frequently need invasive procedures. Numerous mechanisms for thrombocytopenia exist, including splenic sequestration and reduction of levels of the platelet growth factor thrombopoietin. Traditionally, platelet transfusions have been used to increase platelet counts before elective procedures, usually to a threshold of greater than or equal to 50,000/µL, but levels vary by provider, procedure, and specific patient. Recently, the thrombopoietin receptor agonists avatrombopag and lusutrombopag were studied and found efficacious for increasing platelet count in the outpatient setting for select patients with advanced liver disease who need a procedure.


Subject(s)
Hemorrhage/etiology , Liver Diseases/complications , Surgical Procedures, Operative/adverse effects , Thrombocytopenia/etiology , Thrombocytopenia/therapy , Chronic Disease , Cinnamates/therapeutic use , Humans , Platelet Count , Platelet Transfusion , Receptors, Thrombopoietin/agonists , Risk Factors , Thiazoles/therapeutic use , Thiophenes/therapeutic use , Thrombocytopenia/blood , Thrombopoiesis/drug effects
15.
Blood ; 136(15): 1773-1782, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32542378

ABSTRACT

G protein-coupled receptors are critical mediators of platelet activation whose signaling can be modulated by members of the regulator of G protein signaling (RGS) family. The 2 most abundant RGS proteins in human and mouse platelets are RGS10 and RGS18. While each has been studied individually, critical questions remain about the overall impact of this mode of regulation in platelets. Here, we report that mice missing both proteins show reduced platelet survival and a 40% decrease in platelet count that can be partially reversed with aspirin and a P2Y12 antagonist. Their platelets have increased basal (TREM)-like transcript-1 expression, a leftward shift in the dose/response for a thrombin receptor-activating peptide, an increased maximum response to adenosine 5'-diphosphate and TxA2, and a greatly exaggerated response to penetrating injuries in vivo. Neither of the individual knockouts displays this constellation of findings. RGS10-/- platelets have an enhanced response to agonists in vitro, but platelet count and survival are normal. RGS18-/- mice have a 15% reduction in platelet count that is not affected by antiplatelet agents, nearly normal responses to platelet agonists, and normal platelet survival. Megakaryocyte number and ploidy are normal in all 3 mouse lines, but platelet recovery from severe acute thrombocytopenia is slower in RGS18-/- and RGS10-/-18-/- mice. Collectively, these results show that RGS10 and RGS18 have complementary roles in platelets. Removing both at the same time discloses the extent to which this regulatory mechanism normally controls platelet reactivity in vivo, modulates the hemostatic response to injury, promotes platelet production, and prolongs platelet survival.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/genetics , RGS Proteins/genetics , Thrombopoiesis/genetics , Animals , Blood Platelets/drug effects , Cell Survival/genetics , Mice , Mice, Knockout , Phosphorylation , Platelet Activating Factor/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count , RGS Proteins/metabolism , Thrombopoiesis/drug effects
16.
Exp Hematol ; 85: 33-46.e6, 2020 05.
Article in English | MEDLINE | ID: mdl-32417303

ABSTRACT

Thrombopoietin (THPO) and its receptor myeloproliferative leukemia virus oncogene (MPL) regulate hematopoietic stem cell (HSC) quiescence and maintenance, but also megakaryopoiesis. Thrombocytopenias or aplastic anemias can be treated today with THPO peptide mimetics (romiplostim) or small-molecule THPO receptor agonists (e.g., eltrombopag). These THPO mimetics were designed for human application; however, many preclinical studies are performed in murine models. We investigated the activation of wild-type murine MPL (mMPL) by romiplostim. Romiplostim stimulated AKT, ERK1/2, and STAT5 phosphorylation without preference for one of these pathways, however, with a four- to fivefold lower phosphorylation intensity at high concentration. Faster internalization of mMPL after romiplostim binding could be one explanation of reduced signaling. In vitro megakaryocyte differentiation, proliferation, and maturation by romiplostim was less efficient compared with stimulation with mTHPO. We further dissected mMPL signaling by lentiviral overexpression of mMPL mutants with tyrosine (Y)-to-phenylalanine (F) substitutions in the distal cytoplasmic tyrosines 582 (Y582F), 616 (Y616F), and 621 (Y621F) individually and in combination (Y616F_Y621F) and in truncated receptors lacking 53 (Δ53) or 69 (Δ69) C-terminal amino acids. Mutation at tyrosine residue Y582F caused a gain-of-function with baseline activation and increased ERK1/2 phosphorylation upon stimulation. In agreement with this, proliferation in Y582F-32D cells was increased, yet did not rescue in vitro megakaryopoiesis from Mpl-deficient cells. Y616F and Y621F mutated receptors exhibited strongly impaired ERK1/2 and decreased AKT signaling and conferred reduced proliferation to 32D cells upon mTHPO stimulation but a partial correction of immature megakaryopoiesis in vitro.


Subject(s)
MAP Kinase Signaling System/drug effects , Mutation, Missense , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/pharmacology , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology , Amino Acid Substitution , Animals , Cell Line , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Receptors, Fc , Receptors, Thrombopoietin/genetics , Thrombopoiesis/genetics
17.
Biomed Res Int ; 2020: 7636290, 2020.
Article in English | MEDLINE | ID: mdl-32420370

ABSTRACT

A study of myelostimulating activity of ionic compounds-trimecaine alkyl iodide derivatives under the cipher BIV (BIV-117, BIV-118, and BIV-119) was conducted on a model of doxorubicin pancytopenia in white laboratory rats. It was established that the compounds BIV-117 and BIV-119 had a pronounced leukopoiesis-stimulating activity, exceeding the activity of the methyluracil as a comparison drug. Compounds BIV-117 and BIV-119 had erythropoiesis- and thrombocytopoiesis-stimulating activity at the level of methyluracil.


Subject(s)
Leukopoiesis/drug effects , Trimecaine/pharmacology , Animals , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Erythropoiesis/drug effects , Female , Rats , Thrombopoiesis/drug effects
18.
Br J Haematol ; 190(3): 418-429, 2020 08.
Article in English | MEDLINE | ID: mdl-32130729

ABSTRACT

Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is driven by complex genetic and epigenetic alterations from an aberrant clone of hematopoietic stem/progenitor cells (HSPCs). Ubiquitin-specific protease 7 (USP7) has been demonstrated to have an important oncogenic role in the development of several cancer types, but its role in MDS is unknown. Here, we demonstrate that USP7 expression is elevated in MDS cell lines and patient samples. The USP7-selective small-molecule inhibitors P5091 and P22077 inhibited cell proliferation and induced megakaryocytic differentiation in both cell lines and primary cells. Furthermore, pharmacological inhibition of USP7 markedly suppressed the growth of MDS cell lines in xenograft mouse models. To explore the mechanisms underlying the observed phenotypic changes, we employed RNA-seq to compare the differences in genes after USP7 inhibitor treatment and found that gelsolin (GSN) expression was increased significantly after USP7 inhibitor treatment. Furthermore, knockdown of GSN attenuated the proliferation inhibition, apoptosis induction and megakaryocyte differentiation induced by USP7 inhibitors in MDS cells. Collectively, our findings identify previously unknown roles of USP7 and suggest that the USP7/GSN axis may be a potential therapeutic target in MDS.


Subject(s)
Gelsolin/physiology , Megakaryocytes/drug effects , Myelodysplastic Syndromes/pathology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Thrombopoiesis/drug effects , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line/transplantation , Enzyme Induction/drug effects , Gelsolin/biosynthesis , Gelsolin/genetics , Heterografts , Humans , Megakaryocytes/pathology , Mice , Mice, Inbred NOD , Neoplasms, Experimental/etiology , Risk , Transcriptome/drug effects , Ubiquitin-Specific Peptidase 7/physiology , Up-Regulation/drug effects
19.
Blood ; 136(1): 119-129, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32202634

ABSTRACT

Abnormal megakaryocyte development and platelet production lead to thrombocytopenia or thrombocythemia and increase the risk of hemorrhage or thrombosis. Acylglycerol kinase (AGK) is a mitochondrial membrane kinase that catalyzes the formation of phosphatidic acid and lysophosphatidic acid. Mutation of AGK has been described as the major cause of Sengers syndrome, and the patients with Sengers syndrome have been reported to exhibit thrombocytopenia. In this study, we found that megakaryocyte/platelet-specific AGK-deficient mice developed thrombocytopenia and splenomegaly, mainly caused by inefficient bone marrow thrombocytopoiesis and excessive extramedullary hematopoiesis, but not by apoptosis of circulating platelets. It has been reported that the G126E mutation arrests the kinase activity of AGK. The AGK G126E mutation did not affect peripheral platelet counts or megakaryocyte differentiation, suggesting that the involvement of AGK in megakaryocyte development and platelet biogenesis was not dependent on its kinase activity. The Mpl/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (Stat3) pathway is the major signaling pathway regulating megakaryocyte development. Our study confirmed that AGK can bind to JAK2 in megakaryocytes/platelets. More interestingly, we found that the JAK2 V617F mutation dramatically enhanced the binding of AGK to JAK2 and greatly facilitated JAK2/Stat3 signaling in megakaryocytes/platelets in response to thrombopoietin. We also found that the JAK2 JAK homology 2 domain peptide YGVCF617CGDENI enhanced the binding of AGK to JAK2 and that cell-permeable peptides containing YGVCF617CGDENI sequences accelerated proplatelet formation. Therefore, our study reveals critical roles of AGK in megakaryocyte differentiation and platelet biogenesis and suggests that targeting the interaction between AGK and JAK2 may be a novel strategy for the treatment of thrombocytopenia or thrombocythemia.


Subject(s)
Mutation, Missense , Phosphotransferases (Alcohol Group Acceptor)/physiology , Point Mutation , Splenomegaly/genetics , Thrombocytopenia/genetics , Thrombopoiesis/physiology , Amino Acid Sequence , Animals , Blood Platelets/enzymology , Cells, Cultured , Hematopoiesis, Extramedullary/physiology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Liver/cytology , Liver/embryology , Megakaryocytes/enzymology , Mice , Mice, Knockout , Mitochondrial Membranes/enzymology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Interaction Mapping , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Splenomegaly/enzymology , Thrombocytopenia/enzymology , Thrombopoiesis/drug effects
20.
J Pineal Res ; 68(3): e12637, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32052470

ABSTRACT

Melatonin (MT), endogenously secreted by the pineal gland, is closely related to multiple biological processes; however, its effect on thrombopoiesis is still not well illustrated. Here, we demonstrate that MT administration can elevate peripheral platelet levels. Analysis of different stages in thrombopoiesis reveals that MT has the capacity to promote the expansion of CD34+ and CD41+ cells, and accelerate proplatelet formation (PPF) and platelet production. Furthermore, in vivo experiments show that MT has a potential therapeutic effect on radiation-induced thrombocytopenia. The underlying mechanism suggests that both extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt signaling are involved in the processes of thrombopoiesis facilitated by MT. Interestingly, in addition to the direct regulation of Akt signaling by its upstream phosphoinositide 3-kinase (PI3K), ERK1/2 signaling is also regulated by PI3K via its effector, dual adaptor for phosphotyrosine and 3-phosphoinositides (DAPP1), in megakaryocytes after MT treatment. Moreover, the expression level of DAPP1 during megakaryocyte differentiation is closely related to the activation of ERK1/2 and Akt at different stages of thrombopoiesis. In conclusion, our data suggest that MT treatment can promote thrombopoiesis, which is modulated by the DAPP1-orchestrated activation of ERK1/2 and Akt signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MAP Kinase Signaling System/drug effects , Melatonin/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Thrombopoiesis/drug effects , Thrombopoiesis/physiology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Lipoproteins/metabolism , MAP Kinase Signaling System/physiology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...