Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.624
Filtrar
1.
J Biosci ; 492024.
Artigo em Inglês | MEDLINE | ID: mdl-38726826

RESUMO

Lung cancer (LC) is the leading cause of cancer-associated deaths worldwide, among which non-small-cell lung cancer (NSCLC) accounts for 80%. Stromal cell-derived factor-1 (SDF-1) inhibition results in a significant depletion of NSCLC metastasis. Additionally, SDF-1 is the only natural chemokine known to bind and activate the receptor CXCR4. Thus, we attempted to clarify the molecular mechanism of SDF-1 underlying NSCLC progression. Transwell migration, adhesion, and G-LISA assays were used to assess megakaryocytic chemotaxis in vitro and in vivo in terms of megakaryocytic migration, adherence, and RhoA activation, respectively. Western blotting was used to assess PI3K/Akt-associated protein abundances in MEG-01 cells and primary megakaryocytes under the indicated treatment. A hematology analyzer and flow cytometry were used to assess platelet counts in peripheral blood and newly formed platelet counts in Lewis LC mice under different treatments. Immunochemistry and flow cytometry were used to measure CD41+ megakaryocyte numbers in Lewis LC mouse tissue under different treatments. ELISA was used to measure serum TPO levels, and H&E staining was used to detect NSCLC metastasis.SDF-1 receptor knockdown suppressed megakaryocytic chemotaxis in Lewis LC mice. SDF-1 receptor inhibition suppressed megakaryocytic chemotaxis via the PI3K/Akt pathway. SDF-1 receptor knockdown suppressed CD41+ megakaryocyte numbers in vivo through PI3K/Akt signaling. SDF-1 receptor inhibition suppressed CD41+ megakaryocytes to hinder NSCLC metastasis. SDF-1 facilitates NSCLC metastasis by enhancing the chemoattraction of megakaryocytes via the PI3K/Akt signaling pathway, which may provide a potential new direction for seeking therapeutic plans for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Quimiocina CXCL12 , Quimiotaxia , Neoplasias Pulmonares , Megacariócitos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Receptores CXCR4 , Transdução de Sinais , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Megacariócitos/metabolismo , Megacariócitos/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Animais , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/genética , Camundongos , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Linhagem Celular Tumoral , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Metástase Neoplásica , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
2.
J Autoimmun ; 145: 103204, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38520895

RESUMO

Epidemiological studies show that cardiovascular events related to platelet hyperactivity remain the leading causes of death among multiple sclerosis (MS) patients. Quantitative or structural changes of platelet cytoskeleton alter their morphology and function. Here, we demonstrated, for the first time, the structural changes in MS platelets that may be related to their hyperactivity. MS platelets were found to form large aggregates compared to control platelets. In contrast to the control, the images of overactivated, irregularly shaped MS platelets show changes in the cytoskeleton architecture, fragmented microtubule rings. Furthermore, MS platelets have long and numerous pseudopodia rich in actin filaments. We showed that MS platelets and megakaryocytes, overexpress ß1-tubulin and ß-actin mRNAs and proteins and have altered post-translational modification patterns. Moreover, we identified two previously undisclosed mutations in the gene encoding ß1-tubulin in MS. We propose that the demonstrated structural changes of platelet cytoskeleton enhance their ability to adhere, aggregate, and degranulate fueling the risk of adverse cardiovascular events in MS.


Assuntos
Plaquetas , Proteínas do Citoesqueleto , Citoesqueleto , Esclerose Múltipla , Tubulina (Proteína) , Humanos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Esclerose Múltipla/sangue , Plaquetas/metabolismo , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Feminino , Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Adulto , Masculino , Pessoa de Meia-Idade , Actinas/metabolismo , Actinas/genética , Megacariócitos/metabolismo , Megacariócitos/patologia , Processamento de Proteína Pós-Traducional , Mutação
4.
Mil Med Res ; 10(1): 66, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38111039

RESUMO

BACKGROUND: The essential roles of platelets in thrombosis have been well recognized. Unexpectedly, thrombosis is prevalent during thrombocytopenia induced by cytotoxicity of biological, physical and chemical origins, which could be suffered by military personnel and civilians during chemical, biological, radioactive, and nuclear events. Especially, thrombosis is considered a major cause of mortality from radiation injury-induced thrombocytopenia, while the underlying pathogenic mechanism remains elusive. METHODS: A mouse model of radiation injury-induced thrombocytopenia was built by exposing mice to a sublethal dose of ionizing radiation (IR). The phenotypic and functional changes of platelets and megakaryocytes (MKs) were determined by a comprehensive set of in vitro and in vivo assays, including flow cytometry, flow chamber, histopathology, Western blotting, and chromatin immunoprecipitation, in combination with transcriptomic analysis. The molecular mechanism was investigated both in vitro and in vivo, and was consolidated using MK-specific knockout mice. The translational potential was evaluated using a human MK cell line and several pharmacological inhibitors. RESULTS: In contrast to primitive MKs, mature MKs (mMKs) are intrinsically programmed to be apoptosis-resistant through reprogramming the Bcl-xL-BAX/BAK axis. Interestingly, mMKs undergo minority mitochondrial outer membrane permeabilization (MOMP) post IR, resulting in the activation of the cyclic GMP-AMP synthase-stimulator of IFN genes (cGAS-STING) pathway via the release of mitochondrial DNA. The subsequent interferon-ß (IFN-ß) response in mMKs upregulates a GTPase guanylate-binding protein 2 (GBP2) to produce large and hyperreactive platelets that favor thrombosis. Further, we unmask that autophagy restrains minority MOMP in mMKs post IR. CONCLUSIONS: Our study identifies that megakaryocytic mitochondria-cGAS/STING-IFN-ß-GBP2 axis serves as a fundamental checkpoint that instructs the size and function of platelets upon radiation injury and can be harnessed to treat platelet pathologies.


Assuntos
Lesões por Radiação , Trombocitopenia , Trombose , Humanos , Animais , Camundongos , Megacariócitos/metabolismo , Megacariócitos/patologia , Trombocitopenia/etiologia , Apoptose , Nucleotidiltransferases/metabolismo , Trombose/metabolismo
5.
Exp Hematol ; 127: 59-69.e2, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37741606

RESUMO

Most thrombopoietin receptor (MPL) mutations result in abnormal megakaryocyte expansion in the spleen or bone marrow (BM), leading to progressive fibrosis. It has been reported that p21 (Rac Family Small GTPase 1 [RAC1])-activated kinase 1 (PAK1) participates in the proliferation and differentiation of megakaryoblasts. PAK1 phosphorylation increased in patients with myeloproliferative neoplasms (MPNs) and murine MPN cells with the Mplw515l mutant gene in this study; however, the function of overactivated PAK1 in MPN cells remains unclear. We found that inhibition of PAK1 caused significant changes in the biological behaviors of MPLW515L mutant cells in vitro, including arrested growth or reduced clonality and increased polyploid DNA and cell apoptosis due to upregulated cleaved caspase 3. In vivo, PAK1 inhibitor treatment caused a slow elevation of leukocytosis and hematocrit (HCT) and a reduction in hepatosplenomegaly in 6133/MPLW515L-transplanted mice, along with reduced tumor cell infiltration and prolonged survival. Further, deletion of PAK1 sustained a relatively normal HCT and platelet count at the beginning of the disease but did not completely alleviate the splenomegaly of MPLW515L mutant mice. Notably, PAK1 knockout attenuated the destruction of splenic structure, and reduced the megakaryocyte burden within the BM. These results suggest that inhibition of PAK1 may be a useful method for treating MPLW515L mutant MPN by intervening megakaryocytes.


Assuntos
Transtornos Mieloproliferativos , Neoplasias , Humanos , Camundongos , Animais , Megacariócitos/patologia , Proliferação de Células , Neoplasias/patologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Diferenciação Celular , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/farmacologia
6.
Expert Rev Hematol ; 16(10): 731-742, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37668243

RESUMO

INTRODUCTION: Immune thrombocytopenia [ITP] is a common bleeding disorder with an isolated platelet count of less than 100 × 109/L. AREAS COVERED: Relevant literature from 2003 to 2022 was retrieved and reviewed from the Google Scholar search engine and PubMed database. Antibodies produced by autoreactive B lymphocytes and the phagocytic function of macrophages are considered the most critical factors in platelet destruction. Also, macrophages present the antigen to T lymphocytes and activate them. Follicular helper T-cells [TFH] play a role in stimulating, differentiating, and activating autoreactive B cells, while cluster of differentiation [CD]-8+ T plays a role in platelet destruction through apoptosis. The classical pathway of the complement system also causes platelet destruction. By inhibiting platelet production, low levels of thrombopoietin and an immune response against megakaryocytes in the bone marrow worsen thrombocytopenia. EXPERT OPINION: T-cell subset changes and an increase in activated autoreactive B cells, in addition to the function of components of the innate immune system [the complement system, dendritic cells, and natural killer cells], play a critical role in the pathogenesis of the ITP. Accurate detection of these changes may lead to developing new therapeutic strategies and identifying better prognostic/diagnostic factors.


Assuntos
Púrpura Trombocitopênica Idiopática , Trombocitopenia , Humanos , Púrpura Trombocitopênica Idiopática/etiologia , Púrpura Trombocitopênica Idiopática/terapia , Plaquetas , Trombocitopenia/etiologia , Contagem de Plaquetas , Megacariócitos/patologia
7.
Platelets ; 34(1): 2237592, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37577973

RESUMO

Although thrombocytopenia in neonatal intensive care patients is rarely due to inherited disorders, the number of genetic variants implicated in platelet defects has grown dramatically with increasing genome-wide sequencing. Here we describe a case of severe, oligogenic neonatal thrombocytopenia and reinterpret a reportedly benign mutation that is likely pathogenic. Despite this patient's synonymous mutation (GFI1B 576 C>T, Phe192=) being annotated as benign, GFI1B is a well-known regulator of megakaryopoiesis, this variant alters splicing and megakaryocyte maturation, and our analysis of existing genome-wide associated studies demonstrates that it likely causes gray platelet syndrome. This variant has not been reported in a case of life-threatening thrombocytopenia. We propose that the severity of this patient's phenotype is due to synergistic epistasis between the intrinsic platelet defect caused by this mutation and her concomitant inherited PMM2 congenital glycosylation disorder neither of which have been associated with such a severe phenotype. This case highlights the importance of whole-exome/genome sequencing for critically ill patients, reexamining variant interpretation when clinically indicated, and the need to study diverse genetic variation in hematopoiesis.


What is the context? Low platelets (thrombocytopenia) in the neonatal population is not frequently inherited. As we perform unbiased DNA sequencing in more patients, the number of inherited platelet disorders and implicated variants is growing.The gene GFI1B encodes for a transcription factor that regulates megakaryocytes, the cell type that produces platelets. A synonymous substitution in GFI1B (576 C>T, Phe192=) is annotated as benign; however, experimental studies have shown that it inhibits megakaryocyte production.There is growing appreciation for oligogenic inheritance, where multiple causal variants contribute to clinical phenotypes.What is new? We present a case of life-threatening neonatal macrothrombocytopenia (large, hypogranulated sparse platelets) that has an oligogenic cause. We reinterpret the synonymous substitution GFI1B 576 C>T as pathogenic.This patient's severe phenotype was likely due to the combined effect of GFI1B 576 C>T and her inherited glycosylation disorder (PMM2-CDG). Neither variant alone causes severe thrombocytopenia, but the combined intrinsic platelet defect (GFI1B mutation) and consumption (PMM2-CDG) likely produced her life-threatening phenotype.What is the impact? GFI1B is a critical regulator of megakaryocyte production. The purportedly benign mutation 576 C>T is likely pathogenic causing thrombocytopenia by impairing megakaryocyte maturation.As more patients have unbiased genome sequencing, oligogenic and polygenic inheritance will become increasingly appreciated as causes of platelet disorders.NICU providers should consider whole genome or exome sequencing of neonates with severe thrombocytopenia after reversible causes are ruled out.


Assuntos
Trombocitopenia Neonatal Aloimune , Feminino , Humanos , Megacariócitos/patologia , Proteínas Repressoras , Plaquetas/patologia , Mutação , Proteínas Proto-Oncogênicas/genética
8.
Hum Vaccin Immunother ; 19(2): 2246542, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37614152

RESUMO

A good safety and immunogenicity profile was reported in Phase I and II clinical trials of inactivated SARS-CoV-2 vaccines. Here, we report two cases associated with vaccine-associated adverse events, including one patient with fever and another with anaphylactic shock resulting from inactivated SARS-CoV-2 vaccination. Cell sub-types and the importance of genetic characteristics were assessed using single-cell mRNA sequencing and machine learning. Overall, the patient with fever showed a significant increase in the numbers of cytotoxic CD8 T cells and MKI67high CD8 T cells. A potential concurrent infection with the Epstein-Barr virus enhanced interferon type I responses to vaccination against the virus. STAT1, E2F1, YBX1, and E2F7 played a key role in the transcription regulation of MKI67high CD8 T cells. In contrast, the patient with allergic shock displayed predominant increases in the numbers of S100A9high monocytes, activated CD4 T cells, and PPBPhigh megakaryocytes. The decision tree showed that LYZ and S100A8 in S100A9high monocytes contributed to the degranulation of neutrophils and activation of neutrophils involved in allergic shock. PPBP and PF4 were major contributors to platelet degranulation. These findings highlight the diversity of adverse reactions following inactivated SARS-CoV-2 vaccination and show the emerging role of cellular subtypes and central genes in vaccine-associated adverse reactions.


The identification of cell sub-types may help in the diagnosis of COVID-19 vaccine-related adverse events.COVID-19 vaccination-related acute pulmonary edema may induce a higher risk of thrombosis.The long-term fever after vaccination may attribute to the excessive type I interferon responses.


Assuntos
Vacinas contra COVID-19 , Humanos , Masculino , Feminino , Adulto , Vacinas contra COVID-19/efeitos adversos , Febre/imunologia , Febre/patologia , Edema Pulmonar/imunologia , Edema Pulmonar/patologia , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Megacariócitos/patologia , Análise da Expressão Gênica de Célula Única , Linfócitos B/citologia , Monócitos/citologia , Anafilaxia/imunologia , Anafilaxia/patologia
9.
Infect Immun ; 91(8): e0010223, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37404144

RESUMO

Fusobacterium nucleatum colonization contributes to the occurrence of portal vein thrombosis in patients with gastric cancer (GC). However, the underlying mechanism by which F. nucleatum promotes thrombosis remains unclear. In this study, we recruited a total of 91 patients with GC and examined the presence of F. nucleatum in tumor and adjacent non-tumor tissues by fluorescence in situ hybridization and quantitative PCR. Neutrophil extracellular traps (NETs) were detected by immunohistochemistry. Extracellular vesicles (EVs) were extracted from the peripheral blood and proteins in the EVs were identified by mass spectrometry (MS). HL-60 cells differentiated into neutrophils were used to package engineered EVs to imitate the EVs released from NETs. Hematopoietic progenitor cells (HPCs) and K562 cells were used for megakaryocyte (MK) in vitro differentiation and maturation to examine the function of EVs. We observed that F. nucleatum-positive patients had increased NET and platelet counts. EVs from F. nucleatum-positive patients could promote the differentiation and maturation of MKs and had upregulated 14-3-3 proteins, especially 14-3-3ε. 14-3-3ε upregulation promoted MK differentiation and maturation in vitro. HPCs and K562 cells could receive 14-3-3ε from the EVs, which interacted with GP1BA and 14-3-3ζ to trigger PI3K-Akt signaling. In conclusion, we identified for the first time that F. nucleatum infection promotes NET formation, which releases EVs containing 14-3-3ε. These EVs could deliver 14-3-3ε to HPCs and promote their differentiation into MKs via activation of PI3K-Akt signaling.


Assuntos
Vesículas Extracelulares , Infecções por Fusobacterium , Neoplasias Gástricas , Humanos , Fusobacterium nucleatum/metabolismo , Hibridização in Situ Fluorescente , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Megacariócitos/metabolismo , Megacariócitos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Infecções por Fusobacterium/metabolismo , Infecções por Fusobacterium/microbiologia , Infecções por Fusobacterium/patologia , Vesículas Extracelulares/metabolismo
10.
Int J Mol Sci ; 24(12)2023 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-37373420

RESUMO

Extracellular microparticles provide a means of cell-to-cell communication and can promote information exchanges between adjacent or distant cells. Platelets are cell fragments that are derived from megakaryocytes. Their main functions are to stop bleeding, regulate inflammation, and maintain the integrity of blood vessels. When platelets are activated, they can perform related tasks by secreting platelet-derived microparticles that contain lipids, proteins, nucleic acids, and even organelles. There are differences in the circulating platelet levels in many autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, antiphospholipid antibody syndrome, and Sjogren's syndrome. In this paper, the latest findings in the research field of platelet-derived microparticles are reviewed, including the potential pathogenesis of platelet-derived microparticles in various types of immune diseases, their potential as related markers, and for monitoring the progress and prognosis of disease treatment are expounded.


Assuntos
Artrite Reumatoide , Doenças Autoimunes , Micropartículas Derivadas de Células , Lúpus Eritematoso Sistêmico , Humanos , Micropartículas Derivadas de Células/metabolismo , Doenças Autoimunes/metabolismo , Plaquetas/metabolismo , Artrite Reumatoide/metabolismo , Megacariócitos/patologia
11.
Biochemistry (Mosc) ; 88(Suppl 1): S39-S51, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37069113

RESUMO

Platelets are formed from bone marrow megakaryocytes, circulate in blood for 7-10 days, and then are destroyed in the spleen and/or liver. Platelet production depends on the megakaryocyte population state in the bone marrow: number and size of the cells. The platelet turnover, i.e., the number of platelets passing through the bloodstream in a certain time, is determined by both the rate of their production and the rate of their destruction. The review considers laboratory markers, which are used to assess platelet production and turnover in the patients with hematologic and cardiovascular pathologies. These markers include some characteristics of platelets themselves: (i) content of reticulated ("young") forms in the blood detected by their staining with RNA dyes; (ii) indicators of the platelet size determined in hematology analyzers (mean volume, percentage of large forms) and in flow cytometers (light scattering level). Alterations of platelet production and turnover lead to the changes in blood plasma concentrations of such molecules as thrombopoietin (TPO, main mediator of megakaryocyte maturation and platelet formation in the bone marrow) and glycocalicin (soluble fragment of the membrane glycoprotein Ib detached from the surface of platelets during their destruction). Specific changes in the markers of platelet production and turnover have been observed in: (i) hypoproductive thrombocytopenias caused by suppression of megakaryocytes in the bone marrow; (ii) immune thrombocytopenias caused by accelerated clearance of the autoantibody-sensitized platelets; and (iii) thrombocytosis (both primary and reactive). The paper presents the data indicating that in patients with cardiovascular diseases an increased platelet turnover and changes in the corresponding markers (platelet size indexes and content of reticulated forms) are associated with the decreased efficacy of antiplatelet drugs and increased risk of thrombotic events, myocardial infarction, and unstable angina (acute coronary syndrome).


Assuntos
Plaquetas , Trombocitopenia , Humanos , Contagem de Plaquetas , Megacariócitos/patologia , Trombocitopenia/etiologia , Trombocitopenia/patologia , Biomarcadores
12.
Leukemia ; 37(4): 725-727, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36871061

RESUMO

In this Perspective, we discuss criteria for defining a new disease entity or variant of a recognized disease or disorder. We do so in the context of the current topography of the BCR::ABL-negative myeloproliferative neoplasms (MPNs) where two new variants are reported: clonal megakaryocyte dysplasia with normal blood values (CMD-NBV) and clonal megakaryocyte dysplasia with isolated thrombocytosis (CMD-IT). The cardinal feature of these variants is bone marrow megakaryocyte hyperplasia and atypia corresponding the WHO histological criteria for primary myelofibrosis (myelofibrosis-type megakaryocyte dysplasia-MTMD). Persons with these new variants have a different disease course and features from others in the MPN domain. In a broader context we suggest myelofibrosis-type megakaryocyte dysplasia defines a spectrum of related MPN variants including CMD-NBV, CMD-IT, pre-fibrotic myelofibrosis and overt myelofibrosis, which differ from polycythemia vera and essential thrombocythemia. Our proposal needs external validation and we stress the need for a consensus definition of the megakaryocyte dysplasia which is the hallmark of these disorders.


Assuntos
Doenças da Medula Óssea , Transtornos Mieloproliferativos , Policitemia Vera , Mielofibrose Primária , Trombocitose , Humanos , Doenças da Medula Óssea/patologia , Megacariócitos/patologia , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Policitemia Vera/patologia , Mielofibrose Primária/genética , Mielofibrose Primária/patologia , Trombocitose/genética , Trombocitose/patologia , Proteínas de Fusão bcr-abl
13.
Blood ; 141(18): 2261-2274, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36790527

RESUMO

Pathogenic missense variants in SLFN14, which encode an RNA endoribonuclease protein that regulates ribosomal RNA (rRNA) degradation, are known to cause inherited thrombocytopenia (TP) with impaired platelet aggregation and adenosine triphosphate secretion. Despite mild laboratory defects, the patients displayed an obvious bleeding phenotype. However, the function of SLFN14 in megakaryocyte (MK) and platelet biology remains unknown. This study aimed to model the disease in an immortalized MK cell line (imMKCL) and to characterize the platelet transcriptome in patients with the SLFN14 K219N variant. MK derived from heterozygous and homozygous SLFN14 K219N imMKCL and stem cells of blood from patients mainly presented with a defect in proplatelet formation and mitochondrial organization. SLFN14-defective platelets and mature MK showed signs of rRNA degradation; however, this was absent in undifferentiated imMKCL cells and granulocytes. Total platelet RNA was sequenced in 2 patients and 19 healthy controls. Differential gene expression analysis yielded 2999 and 2888 significantly (|log2 fold change| >1, false discovery rate <0.05) up- and downregulated genes, respectively. Remarkably, these downregulated genes were not enriched in any biological pathway, whereas upregulated genes were enriched in pathways involved in (mitochondrial) translation and transcription, with a significant upregulation of 134 ribosomal protein genes (RPGs). The upregulation of mitochondrial RPGs through increased mammalian target of rapamycin complex 1 (mTORC1) signaling in SLFN14 K219N MK seems to be a compensatory response to rRNA degradation. mTORC1 inhibition with rapamycin resulted in further enhanced rRNA degradation in SLFN14 K219N MK. Taken together, our study indicates dysregulation of mTORC1 coordinated ribosomal biogenesis is the disease mechanism for SLFN14-related TP.


Assuntos
Trombocitopenia , Humanos , Trombocitopenia/patologia , Plaquetas/metabolismo , Ribossomos/metabolismo , Megacariócitos/patologia , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , RNA/metabolismo
14.
Haematologica ; 108(5): 1385-1393, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36226497

RESUMO

Congenital amegakaryocytic thrombocytopenia (CAMT) is a recessive disorder characterized by severe reduction of megakaryocytes and platelets at birth, which evolves toward bone marrow aplasia in childhood. CAMT is mostly caused by mutations in MPL (CAMT-MPL), the gene encoding the receptor of thrombopoietin (THPO), a crucial cytokine regulating hematopoiesis. CAMT can be also due to mutations affecting the THPO coding region (CAMT-THPO). In a child with the clinical picture of CAMT, we identified the homozygous c.-323C>T substitution, affecting a potential regulatory region of THPO. Although mechanisms controlling THPO transcription are not characterized, bioinformatics and in vitro analysis showed that c.-323C>T prevents the binding of transcription factors ETS1 and STAT4 to the putative THPO promoter, impairing THPO expression. Accordingly, in the proband the serum THPO concentration indicates defective THPO production. Based on these findings, the patient was treated with the THPO-mimetic agent eltrombopag, which induced a significant increase in platelet count and stable remission of bleeding symptoms. Herein, we report a novel pathogenic variant responsible for CAMT and provide new insights into the mechanisms regulating transcription of the THPO gene.


Assuntos
Receptores de Trombopoetina , Trombopoetina , Criança , Recém-Nascido , Humanos , Trombopoetina/farmacologia , Receptores de Trombopoetina/genética , Mutação , Megacariócitos/patologia , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-1/genética , Fator de Transcrição STAT4/genética
15.
Thromb Res ; 231: 170-181, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36058760

RESUMO

Autophagy, the continuous recycling of intracellular building blocks, molecules, and organelles is necessary to preserve cellular function and homeostasis. In this context, it was demonstrated that autophagy plays an important role in megakaryopoiesis, the development and differentiation of hematopoietic progenitor cells into megakaryocytes. Furthermore, in recent years, autophagic proteins were detected in platelets, anucleate cells generated by megakaryocytes, responsible for hemostasis, thrombosis, and a key cell in inflammation and host immune responses. In the last decade studies have indicated the occurrence of autophagy in platelets. Moreover, autophagy in platelets was subsequently demonstrated to be involved in platelet aggregation, adhesion, and thrombus formation. Here, we review the current knowledge about autophagy in platelets, its function, and clinical implications. However, at the advent of platelet autophagy research, additional discoveries derived from evolving work will be required to precisely define the contributions of autophagy in platelets, and to expand the ever increasing physiologic and pathologic roles these remarkable and versatile blood cells play.


Assuntos
Plaquetas , Trombose , Humanos , Plaquetas/metabolismo , Megacariócitos/patologia , Trombopoese , Trombose/patologia , Autofagia , Biologia
16.
Klin Padiatr ; 234(6): 388-390, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36379227

RESUMO

Congenital amegakaryocytic thrombocytopenia (CAMT) is an autosomal recessive disorder characterized by severe thrombocytopenia that presents soon after birth and is usually not accompanied by specific somatic malformations [Germeshausen M, Ballmaier M. Best Pract Res Clin Haematol 2021; 34: 101286]. CAMT is more prevalent in females than males [Ballmaier M, Germeshausen M. Semin Thromb Hemost 2011; 37: 673-681; Germeshausen M, Ballmaier M. Haematologica 2021; 106: 2439-2448], in contrast to other congenital bone marrow failure syndromes. Patients with CAMT also exhibit cardiac malformations, cerebellar hypoplasia, growth retardation, and a distinctive facial appearance [Yldrm A T, Günes B T, Oymak Y, et al. Blood Coagul Fibrinolysis 2015; 26: 337-341], although it remains unknown whether these are related to CAMT. Mutations in the MPL gene, which encodes the thrombopoietin receptor, are the pathogenetic cause of CAMT [Germeshausen M, Ballmaier M. Haematologica 2021; 106: 2439-2448]. Since thrombopoietin is involved in the maintenance of hematopoietic stem cells and megakaryocyte development [Germeshausen M, Ballmaier M. Best Pract Res Clin Haematol 2021; 34: 101286], CAMT may eventually manifest as a hematopoietic failure. Currently, allogeneic hematopoietic stem cell transplantation (HSCT) is the only cure for CAMT. Human leukocyte antigen (HLA)-matched siblings are the first-choice donors for HSCT because transplantations from matched unrelated donors have a low success rate [King S, Germeshausen M, Strauss G, et al. Br J Haematol 2005; 131: 636-644]. Cancio et al. [Cancio M, Hebert K, Kim S, et al. Transplant Cell Ther 2022; 28: 101 e101-101 e106] reviewed 86 patients treated over 18 years and reported that although HLA-mismatched donors can extend the survival of CAMT patients, HLA-matched donors are preferred. The present report describes the successful treatment of a 3-year-old girl with CAMT using haploidentical allogeneic HSCT from the father, even though he harbored a mutant MPL gene.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Trombocitopenia , Masculino , Feminino , Humanos , Pré-Escolar , Síndrome Congênita de Insuficiência da Medula Óssea , Megacariócitos/patologia , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/terapia
17.
Indian J Pathol Microbiol ; 65(4): 879-885, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36308198

RESUMO

Purpose: Diagnosis of myelodysplastic syndrome (MDS) primarily relies on the detection of morphological dysplasia in bone marrow. It is subjective and many studies have reported lack of interobserver agreement in reporting. Biopsy is preferred specimen for megakaryocyte assessment. We studied 43 bone marrow biopsies from 40 suspected MDS patient having persistent undiagnosed cytopenia. Utility of immunohistochemistry (IHC) with CD61 and p53 in detecting low-grade MDS was analyzed over routine morphology. Method and Results: Total number of megakaryocytes and number of dysplastic megakaryocytes seen on CD61 IHC was significantly higher than that on H and E stain (P value < 0.05) Out of total 43 biopsies, 13 [30.2%] cases showed dysplastic megakaryocytes that were confirmed by interobserver agreement after IHC. From 30 cases with no significant dysplasia on morphology, 21/43 [48.8%] cases showed >10% dysplastic megakaryocytes on CD61 (P value 0.0001). Nine cases showed no significant dysmegakaryopoiesis with either H and E or CD61 IHC. Fourteen cases could meet higher cut off (30%) of dysmegakaryopoiesis with CD 61 IHC. Out of total 34 cases showing significant dysplasia 7 cases (20.6%) showed positivity for p53 on IHC, which is little less than that reported in low-grade MDS. Conclusion: CD61 IHC is helpful in making correct diagnosis of MDS in cases with minimal dysplasia and should be performed before excluding possibility of MDS on morphology in a patient with undiagnosed cytopenia. IHC is cost effective tool for MDS diagnosis in developing world where access to extensive flow cytometery and molecular testing is limited.


Assuntos
Síndromes Mielodisplásicas , Proteína Supressora de Tumor p53 , Humanos , Imuno-Histoquímica , Proteína Supressora de Tumor p53/análise , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/patologia , Medula Óssea/patologia , Megacariócitos/química , Megacariócitos/patologia , Biomarcadores/análise
18.
Pathol Res Pract ; 237: 154060, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35986964

RESUMO

It has been postulated that platelets are produced by fragmentation of the megakaryocytes within the pulmonary circulation rather than budding of their cytoplasm within the bone marrow. Although literature is scarce depicting the levels of the megakaryocytes within the lungs from previously healthy individuals, there are several studies describing the presence of these cells in human necropsy specimens, and it has been hypothesized that their rearrangements could contribute to the pathogenesis of chronic pulmonary vascular disorders. The objective of this study was to describe the characteristics, distribution and total count of megakaryocytes in explants from lung transplant (LTx) recipients based on the final clinicopathological diagnosis, as well as in samples from LTx donors without previously known pulmonary disease. Using the immunohistochemical marker CD61 we quantified and characterized such cells in 20 biopsy samples from LTx donors and in 30 biopsy samples from LTx recipients with different pathologic conditions: vascular disorders of the lungs, obstructive pulmonary disorders and fibrotic lung diseases. Patients suffering from idiopathic pulmonary arterial hypertension (IPAH) showed morphological differences and strikingly higher numbers of the lungs megakaryocytes (264.5 cells/cm2) compared to all the other groups (the average count among donors was 33.55 megakaryocytes/cm2). Such finding could contribute to the understanding of the origin of vasoconstriction, thrombosis and vascular remodeling of the pulmonary circulation - all the basic mechanisms leading to the development of IPAH, as for there is an increasing evidence of several products of platelets and megakaryocytes to be capable of triggering such processes.


Assuntos
Pneumopatias , Megacariócitos , Humanos , Megacariócitos/patologia , Hipertensão Pulmonar Primária Familiar/patologia , Artéria Pulmonar/patologia , Pulmão/patologia , Plaquetas/patologia , Pneumopatias/patologia
19.
Cancer Gene Ther ; 29(11): 1636-1647, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35650393

RESUMO

Megakaryocytes (MK) are mainly derived from bone marrow and are mainly involved in platelet production. Studies have shown that MK derived from bone marrow may have immune function, and that MK from peripheral blood are associated with prostate cancer. Single-cell transcriptome sequencing can help us better understand the heterogeneity and potential function of MK cell populations in bone marrow (BM), peripheral Blood (PB), and cord blood (CB) of healthy and diseased people.We integrated more than 1.2 million single-cell transcriptome data from 132 samples of PB, BM, and CB from healthy individuals and patients from different dataset. We examined the MK (including MK and product of MK) by single-cell RNA sequencing data analysis methods and identification of MK-related protein expression by the Human Protein atlas. We investigate the relationship between the MK subtype and Non-Small Cell Lung Cancer (NSCLC) in 77 non-cancer and 402 NSCLC. We found that MK were widely distributed and the amount of MK in peripheral blood was more than that in bone marrow and there were specificity MK subtypes in peripheral blood. We found classical MK1 with typical MK characteristics and non-classical MK2 closely related to immunity which was the most common subtype in bone marrow and cord blood. Classical MK1 was closely related to Non-Small Cell Lung Cancer (NSCLC) and can be used as a diagnostic marker. MK2 may have potential adaptive immune function and play a role in tumor NSCLC and autoimmune diseases Systemic Lupus Erythematosus. MK have 14 subtypes and are widely distributed in PB, CB, and BM. MK subtypes are closely related to immunity and have potential to be a diagnostic indicator of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Masculino , Megacariócitos/patologia , Sangue Fetal , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Medula Óssea , Células da Medula Óssea/patologia , Neoplasias Pulmonares/patologia , Análise de Sequência de RNA
20.
J Ayub Med Coll Abbottabad ; 34(2): 389-391, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35576311

RESUMO

Essential thrombocytopenia is the myeloproliferative neoplasm associated with the JAK2/CALR/MPL mutation. It is characterized by an increase in thrombocytes and abnormal megakaryocytes. WHO established the diagnostic criteria for diagnosing the myeloproliferative disorder, which is the combination of molecular, clinical, and histological findings. The appearance of megakaryocytes on bone marrow biopsy is the distinguishing feature to identify myeloproliferative neoplasm, and this short review would like to emphasize the presentation of megakaryocytes in bone marrow biopsy.


Assuntos
Transtornos Mieloproliferativos , Neoplasias , Trombocitemia Essencial , Calreticulina/genética , Humanos , Megacariócitos/patologia , Mutação , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Neoplasias/patologia , Trombocitemia Essencial/diagnóstico , Trombocitemia Essencial/genética , Trombocitemia Essencial/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...