Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
1.
Circ Res ; 135(4): 540-549, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39088641

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.


Subject(s)
Blood Platelets , Megakaryocytes , Thrombopoiesis , Humans , Blood Platelets/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Animals , Thrombopoiesis/physiology
3.
Elife ; 132024 May 17.
Article in English | MEDLINE | ID: mdl-38757931

ABSTRACT

Erythropoiesis and megakaryopoiesis are stringently regulated by signaling pathways. However, the precise molecular mechanisms through which signaling pathways regulate key transcription factors controlling erythropoiesis and megakaryopoiesis remain partially understood. Herein, we identified heat shock cognate B (HSCB), which is well known for its iron-sulfur cluster delivery function, as an indispensable protein for friend of GATA 1 (FOG1) nuclear translocation during erythropoiesis of K562 human erythroleukemia cells and cord-blood-derived human CD34+CD90+hematopoietic stem cells (HSCs), as well as during megakaryopoiesis of the CD34+CD90+HSCs. Mechanistically, HSCB could be phosphorylated by phosphoinositol-3-kinase (PI3K) to bind with and mediate the proteasomal degradation of transforming acidic coiled-coil containing protein 3 (TACC3), which otherwise detained FOG1 in the cytoplasm, thereby facilitating FOG1 nuclear translocation. Given that PI3K is activated during both erythropoiesis and megakaryopoiesis, and that FOG1 is a key transcription factor for these processes, our findings elucidate an important, previously unrecognized iron-sulfur cluster delivery independent function of HSCB in erythropoiesis and megakaryopoiesis.


Subject(s)
Erythropoiesis , Phosphatidylinositol 3-Kinases , Transcription Factors , Humans , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Erythropoiesis/physiology , Hematopoietic Stem Cells/metabolism , HSC70 Heat-Shock Proteins/metabolism , K562 Cells , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Signal Transduction , Thrombopoiesis/physiology , Transcription Factors/metabolism , Transcription Factors/genetics
4.
Scand J Clin Lab Invest ; 84(2): 91-96, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38420711

ABSTRACT

BACKGROUND: The Immature Platelet Fraction (IPF) is an indicator of thrombopoiesis which is a useful parameter in thrombocytopenia. It demonstrates compensatory mechanisms in production of platelets, but currently not implemented in routine clinical practice. The aim of this study was to establish the reproducibility and stability of IPF, for both percentage (%-IPF) and absolute (A-IPF) measurements.Material/methods: A total of 71 samples, of which 45 for reproducibility and 26 for stability analysis, were assayed for full blood count using the Sysmex XN-10 analyser at room temperature (RT:19-25 °C). For reproducibility analysis, IPF measurements were analysed 11 times by different appraisers using the same sample, while for stability analysis, IPF was measured over fourteen hourly-intervals up to 24 h (n = 21) and then separately extended beyond the point of stability to 72 h (n = 5). RESULTS: Reproducibility analysis of %-IPF and A-IPF (n = 45) showed very reliable results, with the range of mean CV% values between 1.25-8.90% and 1.70-9.96%, respectively. On the other hand, overall, stability analysis of %-IPF and A-IPF (n = 21) at RT over 24 h showed reliable results, with pooled mean CV% values of 1.32% and 1.43%, respectively, with no significant difference between %-IPF and A-IPF (p = 0.767 and p = 0.821). All %-IPF and A-IPF values had exceeded the set acceptance criterion of stability (CV% ≥ 10.0%) before 72 h. CONCLUSIONS: Overall, %-IPF and A-IPF reproducibility and storage at RT for 24 h predominantly demonstrates the suitability of their usage for testing on the Sysmex XN-series analysers.


Subject(s)
Blood Platelets , Humans , Reproducibility of Results , Blood Platelets/cytology , Platelet Count/instrumentation , Platelet Count/methods , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Thrombopoiesis/physiology
5.
Cells ; 12(23)2023 12 04.
Article in English | MEDLINE | ID: mdl-38067194

ABSTRACT

The bone marrow (BM) hematopoietic system (HS) gives rise to blood cells originating from hematopoietic stem cells (HSCs), including megakaryocytes (MKs) and red blood cells (erythrocytes; RBCs). Many steps of the cell-fate decision remain to be elucidated, being important for cancer treatment. To explore the role of Wnt/ß-catenin for MK and RBC differentiation, we activated ß-catenin signaling in platelet-derived growth factor b (Pdgfb)-expressing cells of the HS using a Cre-lox approach (Ctnnb1BM-GOF). FACS analysis revealed that Pdgfb is mainly expressed by megakaryocytic progenitors (MKPs), MKs and platelets. Recombination resulted in a lethal phenotype in mutants (Ctnnb1BM-GOFwt/fl, Ctnnb1BM-GOFfl/fl) 3 weeks after tamoxifen injection, showing an increase in MKs in the BM and spleen, but no pronounced anemia despite reduced erythrocyte counts. BM transplantation (BMT) of Ctnnb1BM-GOF BM into lethally irradiated wildtype recipients (BMT-Ctnnb1BM-GOF) confirmed the megakaryocytic, but not the lethal phenotype. CFU-MK assays in vitro with BM cells of Ctnnb1BM-GOF mice supported MK skewing at the expense of erythroid colonies. Molecularly, the runt-related transcription factor 1 (RUNX1) mRNA, known to suppress erythropoiesis, was upregulated in Ctnnb1BM-GOF BM cells. In conclusion, ß-catenin activation plays a key role in cell-fate decision favoring MK development at the expense of erythroid production.


Subject(s)
Megakaryocytes , Thrombopoiesis , beta Catenin , Animals , Mice , beta Catenin/metabolism , Megakaryocyte-Erythroid Progenitor Cells , Megakaryocytes/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Thrombopoiesis/physiology
6.
J Transl Med ; 21(1): 540, 2023 08 12.
Article in English | MEDLINE | ID: mdl-37573325

ABSTRACT

BACKGROUND: Cyclooxygenase (COX)-2 is a rate-limiting enzyme in the biosynthesis of prostanoids, which is mostly inducible by inflammatory cytokines. The participation of COX-2 in the maturation of megakaryocytes has been reported but barely studied in primary immune thrombocytopenia (ITP). METHODS: The expressions of COX-2 and Caspase-1, Caspase-3 and Caspase-3 p17 subunit in platelets from ITP patients and healthy controls (HC), and the expressions of COX-2 and CD41 in bone marrow (BM) of ITP patients were measured and analyzed for correlations. The effects of COX-2 inhibitor on megakaryopoiesis and thrombopoiesis were assessed by in vitro culture of Meg01 cells and murine BM-derived megakaryocytes and in vivo experiments of passive ITP mice. RESULTS: The expression of COX-2 was decreased and Caspase-1 and Caspase-3 p17 were increased in platelets from ITP patients compared to HC. In platelets from ITP patients, the COX-2 expression was positively correlated with platelet count and negatively correlated to the expression of Caspase-1. In ITP patients BM, the expression of CD41 was positively correlated with the expression of COX-2. COX-2 inhibitor inhibited the count of megakaryocytes and impaired the maturation and platelet production in Meg01 cells and bone marrow-derived megakaryocytes. COX-2 inhibitor aggravated thrombocytopenia and damaged megakaryopoiesis in ITP murine model. CONCLUSION: COX-2 plays a vital role in the physiologic and pathologic conditions of ITP by intervening the survival of platelets and impairing the megakaryopoiesis and thrombopoiesis of megakaryocytes.


Subject(s)
Purpura, Thrombocytopenic, Idiopathic , Thrombopoiesis , Animals , Mice , Blood Platelets/metabolism , Caspase 3/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors , Megakaryocytes/metabolism , Thrombopoiesis/physiology
7.
J Thromb Haemost ; 21(11): 3207-3223, 2023 11.
Article in English | MEDLINE | ID: mdl-37336437

ABSTRACT

BACKGROUND: Glucocorticoids are widely known for their immunomodulatory action. Their synthetic analogs are used to treat several autoimmune diseases, including immune thrombocytopenia. However, their efficacy and mechanisms of action in immune thrombocytopenia are not fully understood. OBJECTIVES: To investigate the mechanism of glucocorticoid actions on platelet production. METHODS: The actions of glucocorticoids on platelet production were studied combining in vivo, ex vivo and in vitro approaches. RESULTS: Dexamethasone reduced bleeding in mice and rapidly increased circulating young platelet counts. In vitro glucocorticoid treatment stimulated proplatelet formation by megakaryocytes and platelet-like particle release. This effect was blocked by glucocorticoid receptor antagonist RU486, indicating a glucocorticoid receptor-dependent mechanism. Genome-wide analysis revealed that dexamethasone regulates the expression of >1000 genes related to numerous cellular functions, including predominant cytoplasm and cytoskeleton reorganization. Dexamethasone and other glucocorticoids induced the expression of Gda (the gene encoding guanine deaminase), which has been reported to have a role in dendrite development. Inhibition of guanine deaminase enzymatic activity blocked dexamethasone stimulation of proplatelet formation, implicating a critical role for this enzyme in glucocorticoid-mediated platelet production. CONCLUSION: Our findings identify glucocorticoids as new regulators of thrombopoiesis.


Subject(s)
Guanine Deaminase , Purpura, Thrombocytopenic, Idiopathic , Thrombocytopenia , Mice , Animals , Megakaryocytes/metabolism , Thrombopoiesis/physiology , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Guanine Deaminase/metabolism , Transcriptome , Blood Platelets/metabolism , Thrombocytopenia/metabolism , Dexamethasone/pharmacology
8.
Nat Commun ; 14(1): 2093, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37055385

ABSTRACT

Thrombocytopenia is a major complication in a subset of patients with multiple myeloma (MM). However, little is known about its development and significance during MM. Here, we show thrombocytopenia is linked to poor prognosis in MM. In addition, we identify serine, which is released from MM cells into the bone marrow microenvironment, as a key metabolic factor that suppresses megakaryopoiesis and thrombopoiesis. The impact of excessive serine on thrombocytopenia is mainly mediated through the suppression of megakaryocyte (MK) differentiation. Extrinsic serine is transported into MKs through SLC38A1 and downregulates SVIL via SAM-mediated tri-methylation of H3K9, ultimately leading to the impairment of megakaryopoiesis. Inhibition of serine utilization or treatment with TPO enhances megakaryopoiesis and thrombopoiesis and suppresses MM progression. Together, we identify serine as a key metabolic regulator of thrombocytopenia, unveil molecular mechanisms governing MM progression, and provide potential therapeutic strategies for treating MM patients by targeting thrombocytopenia.


Subject(s)
Multiple Myeloma , Thrombocytopenia , Humans , Bone Marrow/metabolism , Thrombopoiesis/physiology , Multiple Myeloma/complications , Multiple Myeloma/metabolism , Thrombocytopenia/metabolism , Bone Marrow Cells/metabolism , Megakaryocytes , Blood Platelets/metabolism , Tumor Microenvironment
9.
J Thromb Haemost ; 21(2): 344-358, 2023 02.
Article in English | MEDLINE | ID: mdl-36700501

ABSTRACT

BACKGROUND: Platelet shedding from mature megakaryocytes (MKs) in thrombopoiesis is the critical step for elevating circulating platelets fast and efficiently, however, the underlying mechanism is still not well-illustrated, and the therapeutic targets and candidates are even less. OBJECTIVES: In order to investigate the mechanisms for platelet shedding after vasopressin treatment and find new therapeutic targets for thrombocytopenia. METHODS: Platelet production was evaluated both in vivo and in vitro after arginine vasopressin (AVP) administration. The underlying biological mechanism of AVP-triggered thrombopoiesis were then investigated by a series of molecular and bioinformatics techniques. RESULTS: it is observed that proplatelet formation and platelet shedding in the final stages of thrombopoiesis promoted by AVP, an endogenous hormone, can quickly increases peripheral platelets. This rapid elevation is thus able to speed up platelet recovery after radiation as expected. The mechanism analysis reveal that proplatelet formation and platelet release from mature MKs facilitated by AVP is mainly mediated by Akt-regulated mitochondrial metabolism. In particular, phosphorylated Akt regulates mitochondrial metabolism through driving the association of hexokinase-2 with mitochondrial voltage dependent anion channel-1 in AVP-mediated thrombopoiesis. Further studies suggest that this interaction is stabilized by IκBα, the expression of which is controlled by insulin-regulated membrane aminopeptidase. CONCLUSION: these data demonstrate that phosphorylated Akt-mediated mitochondrial metabolism regulates platelet shedding from MKs in response to AVP, which will provide new therapeutic targets and further drug discovery clues for thrombocytopenia treatment.


Subject(s)
Proto-Oncogene Proteins c-akt , Thrombocytopenia , Humans , Proto-Oncogene Proteins c-akt/metabolism , Blood Platelets/metabolism , Megakaryocytes/metabolism , Thrombopoiesis/physiology , Thrombocytopenia/metabolism , Vasopressins/pharmacology , Vasopressins/metabolism
10.
Haematologica ; 108(5): 1394-1411, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36546424

ABSTRACT

Thrombocytopenia is a thrombopoietin (TPO)-related disorder with very limited treatment options, and can be lifethreatening. There are major problems with typical thrombopoietic agents targeting TPO signaling, so it is urgent to discover a novel TPO-independent mechanism involving thrombopoiesis and potential druggable targets. We developed a drug screening model by the multi-grained cascade forest (gcForest) algorithm and found that 3,8-di-O-methylellagic acid 2- O-glucoside (DMAG) (10, 20 and 40 µM) promoted megakaryocyte differentiation in vitro. Subsequent investigations revealed that DMAG (40 mM) activated ERK1/2, HIF-1b and NF-E2. Inhibition of ERK1/2 blocked megakaryocyte differentiation and attenuated the upregulation of HIF-1b and NF-E2 induced by DMAG. Megakaryocyte differentiation induced by DMAG was inhibited via knockdown of NF-E2. In vivo studies showed that DMAG (5 mg/kg) accelerated platelet recovery and megakaryocyte differentiation in mice with thrombocytopenia. The platelet count of the DMAG-treated group recovered to almost 72% and 96% of the count in the control group at day 10 and 14, respectively. The platelet counts in the DMAG-treated group were almost 1.5- and 1.3-fold higher compared with those of the irradiated group at day 10 and 14, respectively. Moreover, DMAG (10, 25 and 50 mM) stimulated thrombopoiesis in zebrafish. DMAG (5 mg/kg) could also increase platelet levels in c-MPL knockout (c-MPL-/-) mice. In summary, we established a drug screening model through gcForest and demonstrated that DMAG promotes megakaryocyte differentiation via the ERK/HIF1/NF-E2 pathway which, importantly, is independent of the classical TPO/c-MPL pathway. The present study may provide new insights into drug discovery for thrombopoiesis and TPO-independent regulation of thrombopoiesis, as well as a promising avenue for thrombocytopenia treatment.


Subject(s)
Anemia , Thrombocytopenia , Animals , Mice , Anemia/metabolism , Blood Platelets/metabolism , Megakaryocytes/metabolism , Thrombocytopenia/metabolism , Thrombopoiesis/physiology , Thrombopoietin/therapeutic use , Zebrafish/metabolism , Glucosides/therapeutic use
11.
Nat Cardiovasc Res ; 2(8): 746-763, 2023 Aug.
Article in English | MEDLINE | ID: mdl-39195958

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.


Subject(s)
Blood Platelets , CD36 Antigens , Fatty Acids, Unsaturated , Megakaryocytes , Phospholipids , Thrombopoiesis , Animals , Blood Platelets/metabolism , Thrombopoiesis/physiology , CD36 Antigens/metabolism , CD36 Antigens/genetics , Phospholipids/metabolism , Megakaryocytes/metabolism , Megakaryocytes/cytology , Humans , Fatty Acids, Unsaturated/metabolism , Mice, Knockout , Thrombocytopenia/metabolism , Male , Mice, Inbred C57BL , Platelet Count , Cells, Cultured , Female , Mice , Lipidomics , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/cytology
12.
Nat Commun ; 13(1): 4504, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35922411

ABSTRACT

Hematopoietic stem cells (HSCs) produce highly diverse cell lineages. Here, we chart native lineage pathways emanating from HSCs and define their physiological regulation by computationally integrating experimental approaches for fate mapping, mitotic tracking, and single-cell RNA sequencing. We find that lineages begin to split when cells leave the tip HSC population, marked by high Sca-1 and CD201 expression. Downstream, HSCs either retain high Sca-1 expression and the ability to generate lymphocytes, or irreversibly reduce Sca-1 level and enter into erythro-myelopoiesis or thrombopoiesis. Thrombopoiesis is the sum of two pathways that make comparable contributions in steady state, a long route via multipotent progenitors and CD48hi megakaryocyte progenitors (MkPs), and a short route from HSCs to developmentally distinct CD48-/lo MkPs. Enhanced thrombopoietin signaling differentially accelerates the short pathway, enabling a rapid response to increasing demand. In sum, we provide a blueprint for mapping physiological differentiation fluxes from HSCs and decipher two functionally distinct pathways of native thrombopoiesis.


Subject(s)
Hematopoietic Stem Cells , Thrombopoiesis , Cell Differentiation/physiology , Cell Lineage , Hematopoietic Stem Cells/metabolism , Myelopoiesis , Thrombopoiesis/physiology
13.
Blood ; 139(17): 2673-2690, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35245376

ABSTRACT

The process of proplatelet formation (PPF) requires coordinated interaction between megakaryocytes (MKs) and the extracellular matrix (ECM), followed by a dynamic reorganization of the actin and microtubule cytoskeleton. Localized fluxes of intracellular calcium ions (Ca2+) facilitate MK-ECM interaction and PPF. Glutamate-gated N-methyl-D-aspartate receptor (NMDAR) is highly permeable to Ca2+. NMDAR antagonists inhibit MK maturation ex vivo; however, there are no in vivo data. Using the Cre-loxP system, we generated a platelet lineage-specific knockout mouse model of reduced NMDAR function in MKs and platelets (Pf4-Grin1-/- mice). Effects of NMDAR deletion were examined using well-established assays of platelet function and production in vivo and ex vivo. We found that Pf4-Grin1-/- mice had defects in megakaryopoiesis, thrombopoiesis, and platelet function, which manifested as reduced platelet counts, lower rates of platelet production in the immune model of thrombocytopenia, and prolonged tail bleeding time. Platelet activation was impaired to a range of agonists associated with reduced Ca2+ responses, including metabotropic like, and defective platelet spreading. MKs showed reduced colony and proplatelet formation. Impaired reorganization of intracellular F-actin and α-tubulin was identified as the main cause of reduced platelet function and production. Pf4-Grin1-/- MKs also had lower levels of transcripts encoding crucial ECM elements and enzymes, suggesting NMDAR signaling is involved in ECM remodeling. In summary, we provide the first genetic evidence that NMDAR plays an active role in platelet function and production. NMDAR regulates PPF through a mechanism that involves MK-ECM interaction and cytoskeletal reorganization. Our results suggest that NMDAR helps guide PPF in vivo.


Subject(s)
Megakaryocytes/metabolism , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Thrombocytopenia , Actins/metabolism , Animals , Blood Platelets/metabolism , Calcium , Mice , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/genetics , Thrombocytopenia/genetics , Thrombopoiesis/physiology
14.
Int J Hematol ; 115(3): 310-321, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35106701

ABSTRACT

C-type lectin-like receptor 2 (CLEC-2) expressed on megakaryocytes plays important roles in megakaryopoiesis. We found that CLEC-2 was expressed in about 20% of phenotypical long-term hematopoietic stem cells (LT-HSCs), which expressed lower levels of HSC-specific genes and produced larger amounts of megakaryocyte-related molecules than CLEC-2low LT-HSCs. Although CLEC-2high LT-HSCs had immature clonogenic activity, cultured CLEC-2high LT-HSCs preferentially differentiated into megakaryocytes. CLEC-2high HSCs yielded 6.8 times more megakaryocyte progenitors (MkPs) and 6.0 times more platelets 2 weeks and 1 week after transplantation compared with CLEC-2low LT-HSCs. However, platelet yield from CLEC-2high HSCs gradually declined with the loss of MkPs, while CLEC-2low HSCs self-renewed long-term, indicating that CLEC-2high LT-HSCs mainly contribute to early megakaryopoiesis. Treatment with pI:C and LPS increased the proportion of CLEC-2high LT-HSCs within LT-HSCs. Almost all CLEC-2low LT-HSCs were in the G0 phase and barely responded to pI:C. In contrast, 54% of CLEC-2high LT-HSCs were in G0, and pI:C treatment obliged CLEC-2high LT-HSCs to enter the cell cycle and differentiate into megakaryocytes, indicating that CLEC-2high LT-HSCs are primed for cell cycle entry and rapidly yield platelets in response to inflammatory stress. In conclusion, CLEC-2high LT-HSCs appear to act as a reserve for emergent platelet production under stress conditions.


Subject(s)
Hematopoietic Stem Cells/physiology , Lectins, C-Type/physiology , Megakaryocytes/metabolism , Thrombopoiesis/genetics , Thrombopoiesis/physiology , Animals , Blood Platelets , Cell Cycle , Cell Differentiation/genetics , Gene Expression , Hematopoietic Stem Cells/metabolism , Inflammation , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice, Inbred C57BL , Phenotype
15.
Blood ; 139(19): 2958-2971, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35176139

ABSTRACT

Translation is essential for megakaryocyte (MK) maturation and platelet production. However, how the translational pathways are regulated in this process remains unknown. In this study, we found that MK/platelet-specific lactate dehydrogenase A (LdhA) knockout mice exhibited an increased number of platelets with remarkably accelerated MK maturation and proplatelet formation. Interestingly, the role of LDHA in MK maturation and platelet formation did not depend on lactate content, which was the major product of LDHA. Mechanism studies revealed that LDHA interacted with eukaryotic elongation factor 2 (eEF2) in the cytoplasm, controlling the participation of eEF2 in translation at the ribosome. Furthermore, the interaction of LDHA and eEF2 was dependent on nicotinamide adenine dinucleotide (NADH), a coenzyme of LDHA. NADH-competitive inhibitors of LDHA could release eEF2 from the LDHA pool, upregulate translation, and enhance MK maturation in vitro. Among LDHA inhibitors, stiripentol significantly promoted the production of platelets in vivo under a physiological state and in the immune thrombocytopenia model. Moreover, stiripentol could promote platelet production from human cord blood mononuclear cell-derived MKs and also have a superposed effect with romiplostim. In short, this study shows a novel nonclassical function of LDHA in translation that may serve as a potential target for thrombocytopenia therapy.


Subject(s)
Elongation Factor 2 Kinase , L-Lactate Dehydrogenase , Megakaryocytes , Thrombocytopenia , Thrombopoiesis , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Elongation Factor 2 Kinase/blood , Elongation Factor 2 Kinase/metabolism , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Knockout , NAD/metabolism , Peptide Elongation Factor 2/metabolism , Thrombocytopenia/blood , Thrombocytopenia/drug therapy , Thrombocytopenia/enzymology , Thrombocytopenia/metabolism , Thrombopoiesis/physiology
16.
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
17.
Indian J Pathol Microbiol ; 64(2): 347-350, 2021.
Article in English | MEDLINE | ID: mdl-33851632

ABSTRACT

Immature platelet fraction (IPF) is a quantification of immature platelets in the circulation reflecting the state of thrombopoiesis in the marrow. Normal reference range for IPF has been established in adults. Reference intervals in neonates are highly dependent on gestational age of the neonate. Complete blood counts (CBC) with IPF of all neonates admitted in neonatal intensive care unit (NICU) were analyzed using Mindray BC-6800 Auto Hematology analyzer. Platelet count of less than 150 × 10^9/L was assigned as thrombocytopenia. Neonates were divided into four groups as per the corrected gestational age (CGA) on the day of CBC analysis: 28-32 weeks, 32-34 weeks, 34-37 weeks, and >37 weeks according to World Health Organization (WHO) classification. Mean, standard deviation, and 95% confidence interval for IPF was calculated in each group and reference range for IPF was derived. Mean IPF in neonates with normal platelet count was term--3.58 (95% CI 3.29 to 3.87), late preterm Neonates (34-37 weeks)--4.14 (95% CI 3.82 to 5.0), moderate preterm neonates (32-34 weeks)--4.14 (95% CI 3.46 to 4.82), and in Very Preterm neonates (28-32 weeks)--IPF of 5.51 (95% CI 3.95 to 7.07). We aimed to establish a reference range for IPF in neonates of different gestational age groups. The IPF values in neonates were comparable between hematology analyzers in neonates with normal platelet counts.


Subject(s)
Blood Platelets/cytology , Blood Platelets/physiology , Thrombocytopenia/diagnosis , Gestational Age , Humans , Infant , Infant, Newborn , Intensive Care Units, Neonatal , Platelet Count , Reference Standards , Reference Values , Thrombopoiesis/physiology
18.
Stem Cells ; 39(6): 787-802, 2021 06.
Article in English | MEDLINE | ID: mdl-33544938

ABSTRACT

Metabolic state of hematopoietic stem cells (HSCs) is an important regulator of self-renewal and lineage-specific differentiation. Posttranslational modification of proteins via O-GlcNAcylation is an ideal metabolic sensor, but how it contributes to megakaryopoiesis and thrombopoiesis remains unknown. Here, we reveal for the first time that cellular O-GlcNAcylation levels decline along the course of megakaryocyte (MK) differentiation from human-derived hematopoietic stem and progenitor cells (HSPCs). Inhibition of O-GlcNAc transferase (OGT) that catalyzes O-GlcNAcylation prolongedly decreases O-GlcNAcylation and induces the acquisition of CD34+ CD41a+ MK-like progenitors and its progeny CD34- CD41a+ /CD42b+ megakaryoblasts (MBs)/MKs from HSPCs, consequently resulting in increased CD41a+ and CD42b+ platelets. Using correlation and co-immunoprecipitation analyses, we further identify c-Myc as a direct downstream target of O-GlcNAcylation in MBs/MKs and provide compelling evidence on the regulation of platelets by novel O-GlcNAc/c-Myc axis. Our data indicate that O-GlcNAcylation posttranslationally regulates c-Myc stability by interfering with its ubiquitin-mediated proteasomal degradation. Depletion of c-Myc upon inhibition of OGT promotes platelet formation in part through the perturbation of cell adhesion molecules, that is, integrin-α4 and integrin-ß7, as advised by gene ontology and enrichment analysis for RNA sequencing and validated herein. Together, our findings provide a novel basic knowledge on the regulatory role of O-GlcNAcylation in megakaryopoiesis and thrombopoiesis that could be important in understanding hematologic disorders whose etiology are related to impaired platelet production and may have clinical applications toward an ex vivo platelet production for transfusion.


Subject(s)
Integrins/metabolism , Megakaryocytes/metabolism , Thrombopoiesis/physiology , Blood Platelets/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Protein Processing, Post-Translational/physiology , Thrombopoiesis/genetics , Transcription Factors/metabolism
19.
Br J Haematol ; 192(5): 909-921, 2021 03.
Article in English | MEDLINE | ID: mdl-33528045

ABSTRACT

Lowe syndrome (LS) is an oculocerebrorenal syndrome of Lowe (OCRL1) genetic disorder resulting in a defect of the OCRL protein, a phosphatidylinositol-4,5-bisphosphate 5-phosphatase containing various domains including a Rho GTPase-activating protein (RhoGAP) homology domain catalytically inactive. We previously reported surgery-associated bleeding in patients with LS, suggestive of platelet dysfunction, accompanied with a mild thrombocytopenia in several patients. To decipher the role of OCRL in platelet functions and in megakaryocyte (MK) maturation, we conducted a case-control study on 15 patients with LS (NCT01314560). While all had a drastically reduced expression of OCRL, this deficiency did not affect platelet aggregability, but resulted in delayed thrombus formation on collagen under flow conditions, defective platelet spreading on fibrinogen and impaired clot retraction. We evidenced alterations of the myosin light chain phosphorylation (P-MLC), with defective Rac1 activity and, inversely, elevated active RhoA. Altered cytoskeleton dynamics was also observed in cultured patient MKs showing deficient proplatelet extension with increased P-MLC that was confirmed using control MKs transfected with OCRL-specific small interfering(si)RNA (siOCRL). Patients with LS also had an increased proportion of circulating barbell-shaped proplatelets. Our present study establishes that a deficiency of the OCRL protein results in a defective actomyosin cytoskeleton reorganisation in both MKs and platelets, altering both thrombopoiesis and some platelet responses to activation necessary to ensure haemostasis.


Subject(s)
Blood Platelets/cytology , Megakaryocytes/cytology , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/physiology , Thrombopoiesis/physiology , Actomyosin/analysis , Adolescent , Adult , Anemia/etiology , Blood Coagulation , Blood Platelets/ultrastructure , Case-Control Studies , Cell Shape , Child , Collagen , Cytoskeleton/ultrastructure , Female , Gene Silencing , Humans , Male , Megakaryocytes/ultrastructure , Middle Aged , Mutation , Myosin Light Chains/metabolism , Oculocerebrorenal Syndrome/blood , Oculocerebrorenal Syndrome/pathology , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Domains , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , Signal Transduction , Thrombocytopenia/etiology , Young Adult
20.
Cytokine ; 147: 155310, 2021 11.
Article in English | MEDLINE | ID: mdl-33127256

ABSTRACT

The blood vascular system of mammals is unique in nature; inhabited with a pool of tiny small cell fragments called platelets; attributed with the most important patrolling tasks to check integrity of the entire endothelial landscape. Their production is tightly coupled with hematopoietic system where everything starts from self renewable multipotent hematopoietic stem cells (HSCs) which eventually undergo dual step (megakaryopoiesis-thrombopoiesis) thrombocytes production. Several cytokines tune the fate of every progenitor cells during hematopoiesis through temporal activation of specific transcription factors. Though platelets generated through steady state hematopoiesis are involved in the regulation of vascular homeostasis, these cells can sense pathogens through its innate immune sensors and can mount crucial responses against the invading pathogen. For this, the primary aim of many infections including Leishmania is to induce thrombocytopenia within infected host. But the underlying mechanism of this induced thrombocytopenia in Leishmania infection has not been evaluated. Elucidation of these mechanisms will be fruitful to design new chemotherapeutic strategies.


Subject(s)
Leishmaniasis/physiopathology , Thrombopoiesis/physiology , Animals , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Immunity, Innate/physiology , Leishmaniasis/metabolism , Thrombocytopenia/metabolism , Thrombocytopenia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL