Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.030
Filtrar
1.
Front Med ; 16(3): 416-428, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34792736

RESUMO

Abivertinib, a third-generation tyrosine kinase inhibitor, is originally designed to target epidermal growth factor receptor (EGFR)-activating mutations. Previous studies have shown that abivertinib has promising antitumor activity and a well-tolerated safety profile in patients with non-small-cell lung cancer. However, abivertinib also exhibited high inhibitory activity against Bruton's tyrosine kinase and Janus kinase 3. Given that these kinases play some roles in the progression of megakaryopoiesis, we speculate that abivertinib can affect megakaryocyte (MK) differentiation and platelet biogenesis. We treated cord blood CD34+ hematopoietic stem cells, Meg-01 cells, and C57BL/6 mice with abivertinib and observed megakaryopoiesis to determine the biological effect of abivertinib on MK differentiation and platelet biogenesis. Our in vitro results showed that abivertinib impaired the CFU-MK formation, proliferation of CD34+ HSC-derived MK progenitor cells, and differentiation and functions of MKs and inhibited Meg-01-derived MK differentiation. These results suggested that megakaryopoiesis was inhibited by abivertinib. We also demonstrated in vivo that abivertinib decreased the number of MKs in bone marrow and platelet counts in mice, which suggested that thrombopoiesis was also inhibited. Thus, these preclinical data collectively suggested that abivertinib could inhibit MK differentiation and platelet biogenesis and might be an agent for thrombocythemia.


Assuntos
Acrilamidas , Plaquetas , Megacariócitos , Piperazinas , Pirimidinas , Acrilamidas/farmacologia , Animais , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Diferenciação Celular , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Piperazinas/farmacologia , Pirimidinas/farmacologia
2.
Sci Rep ; 11(1): 17990, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504210

RESUMO

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.


Assuntos
Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombocitopenia/metabolismo , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Proteína cdc42 de Ligação ao GTP/metabolismo , Actinas/metabolismo , Alquil e Aril Transferases/antagonistas & inibidores , Benzamidas/farmacologia , Plaquetas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células HEK293 , Humanos , Mutação , Prenilação de Proteína/efeitos dos fármacos , Pirazóis/farmacologia , Transdução de Sinais/genética , Sulfonamidas/farmacologia , Síndrome , Trombocitopenia/genética , Trombopoese/efeitos dos fármacos , Trombopoese/genética , Transfecção , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Quinases Ativadas por p21/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo
3.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34153091

RESUMO

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.


Assuntos
16,16-Dimetilprostaglandina E2/uso terapêutico , Síndrome Aguda da Radiação/tratamento farmacológico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Plaquetas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Transtornos Hemorrágicos/tratamento farmacológico , Lisinopril/uso terapêutico , Megacariócitos/efeitos dos fármacos , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Síndrome Aguda da Radiação/complicações , Animais , Plaquetas/efeitos da radiação , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Proteína C-Reativa/análise , Radioisótopos de Césio , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/efeitos da radiação , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/efeitos da radiação , Feminino , Raios gama/efeitos adversos , Transtornos Hemorrágicos/etiologia , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P/análise , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos da radiação , Fator Plaquetário 4/análise , Lesões Experimentais por Radiação/tratamento farmacológico , Lesões Experimentais por Radiação/etiologia , Trombocitopenia/etiologia , Trombopoese/efeitos da radiação , Irradiação Corporal Total , Fator de von Willebrand/análise
4.
Elife ; 102021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34059198

RESUMO

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.


Assuntos
Benzoatos/farmacologia , Plaquetas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hidrazinas/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Pirazóis/farmacologia , Receptores de Trombopoetina/agonistas , Nicho de Células-Tronco , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Adulto , Idoso , Reatores Biológicos , Plaquetas/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Feminino , Fibroínas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Megacariócitos/metabolismo , Pessoa de Meia-Idade , Miniaturização , Mutação , Cadeias Pesadas de Miosina/genética , Receptores de Trombopoetina/metabolismo , Trombocitopenia/sangue , Trombocitopenia/genética , Adulto Jovem
5.
J Ethnopharmacol ; 275: 114074, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33831466

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Carica papaya leaf juice/decoction has been in use in folk medicine in Srilanka, Malaysia and in few parts of India for enhancing the platelet counts in dengue. In Siddha medicine, a traditional form of medicine in India, papaya leaf juice has been used for increasing the platelet counts. Papaya leaf has been reported to enhance blood volume in ancient Ayurveda books in India. Carica papaya leaf is well known for its platelet enhancement activity. Although many preclinical and clinical studies have demonstrated the ability of papaya leaf juice for platelet enhancement, but the underlying mechanisms are still unclear. AIM OF THE STUDY: The study is aimed at identifying the key ingredients of papaya leaf extract and elucidate the mechanism (s) of action of the identified potent component in mitigating thrombocytopenia (Thp). MATERIALS AND METHODS: C. papaya leaf juice was subjected for sequential fractionation to identify the anti-thrombocytopenic phytochemicals. In vivo, stable thrombocytopenia was induced by subcutaneous injection of 70 mg/kg cyclophosphamide (Cyp). After induction, rats were treated with 200 and 400 mg/kg body weight papaya leaf juice and with identified fractions for 14 days. Serum thrombopoietin level was estimated using ELISA. CD110/cMpl, a receptor for thrombopoietin on platelets was measured by western blotting. RESULTS: Administration of cyclophosphamide for 6 days induced thrombocytopenia (210.4 ± 14.2 × 103 cells/µL) in rats. Treating thrombocytopenic rats with papaya leaf juice and butanol fraction for 14 days significantly increased the platelet count to 1073.50 ± 29.6 and 1189.80 ± 36.5 × 103 cells/µL, respectively. C.papaya extracts normalized the elevated bleeding and clotting time and decreased oxidative markers by increasing endogenous antioxidants. A marginal increase in the serum thrombopoietin (TPO) level was observed in Cyp treated group compared to normal and treatment groups. Low expression of CD110/cMpl receptor found in Cyp treated group was enhanced by C. papaya extracts (CPJ) and CPJ-BT. Furthermore, examination of the morphology of bone marrow megakaryocytes, histopathology of liver and kidneys revealed the ability of CPJ and fractions in mitigating Cyp-induced thrombocytopenia in rats. CONCLUSION: C. papaya leaf juice enhances the platelet count in chemotherapy-induced thrombocytopenia by increasing the expression of CD110 receptor on the megakaryocytes. Hence, activating CD110 receptor might be a viable strategy to increase the platelet production in individuals suffering from thrombocytopenia.


Assuntos
Plaquetas/efeitos dos fármacos , Carica/química , Megacariócitos/metabolismo , Extratos Vegetais/farmacologia , Receptores de Trombopoetina/metabolismo , Trombocitopenia/tratamento farmacológico , Administração Oral , Animais , Antioxidantes/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Ciclofosfamida/toxicidade , Modelos Animais de Doenças , Rim/efeitos dos fármacos , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Malondialdeído/metabolismo , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Extratos Vegetais/administração & dosagem , Extratos Vegetais/química , Extratos Vegetais/toxicidade , Folhas de Planta/química , Ratos Sprague-Dawley , Trombocitopenia/sangue , Trombocitopenia/induzido quimicamente , Trombopoetina/sangue
6.
Cancer Immunol Immunother ; 70(12): 3477-3488, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33890137

RESUMO

Acute megakaryocytic leukemia (AMKL) is one of the rarest sub-types of acute myeloid leukemia (AML). AMKL is characterized by high proliferation of megakaryoblasts and myelofibrosis of bone marrow, this disease is also associated with poor prognosis. Previous analyses have reported that the human megakaryoblastic cells can be differentiated into cells with megakaryocyte (MK)-like characteristics by phorbol 12-myristate 13-acetate (PMA). However, little is known about the mechanism responsible for regulating this differentiation process. We performed long non-coding RNA (lncRNA) profiling to investigate the differently expressed lncRNAs in megakaryocyte blast cells treated with and without PMA and examined those that may be responsible for the PMA-induced differentiation of megakaryoblasts into MKs. We found 30 out of 90 lncRNA signatures to be differentially expressed after PMA treatment of megakaryoblast cells, including the highly expressed JPX lncRNA. Further, in silico lncRNA-miRNA and miRNA-mRNA interaction analysis revealed that the JPX is likely involved in unblocking the expression of TGF-ß receptor (TGF-ßR) by sponging oncogenic miRNAs (miR-9-5p, miR-17-5p, and miR-106-5p) during MK differentiation. Further, we report the activation of TGF-ßR-induced non-canonical ERK1/2 and PI3K/AKT pathways during PMA-induced MK differentiation and ploidy development. The present study demonstrates that TGF-ßR-induced non-canonical ERK1/2 and PI3K/AKT pathways are associated with PMA-induced MK differentiation and ploidy development; in this molecular mechanism, JPX lncRNA could act as a decoy for miR-9-5p, miR-17-5p, and miR-106-5p, titrating them away from TGF-ßR mRNAs. Importantly, this study reveals the activation of ERK1/2 and PI3K/AKT pathway in PMA-induced Dami cell differentiation into MK. The identified differentially expressed lncRNA signatures may facilitate further study of the detailed molecular mechanisms associated with MK development. Thus, our data provide numerous targets with therapeutic potential for the modulation of the differentiation of megakaryoblastic cells in AMKL.


Assuntos
Leucemia Megacarioblástica Aguda/tratamento farmacológico , Megacariócitos/efeitos dos fármacos , Ésteres de Forbol/farmacologia , RNA Longo não Codificante/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Leucemia Megacarioblástica Aguda/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA Longo não Codificante/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética
7.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33804889

RESUMO

In chronic kidney disease, hyperphosphatemia upregulates the Ca2+ channel ORAI and its activating Ca2+ sensor STIM in megakaryocytes and platelets. ORAI1 and STIM1 accomplish store-operated Ca2+ entry (SOCE) and play a key role in platelet activation. Signaling linking phosphate to upregulation of ORAI1 and STIM1 includes transcription factor NFAT5 and serum and glucocorticoid-inducible kinase SGK1. In vascular smooth muscle cells, the effect of hyperphosphatemia on ORAI1/STIM1 expression and SOCE is suppressed by Mg2+ and the calcium-sensing receptor (CaSR) agonist Gd3+. The present study explored whether sustained exposure to Mg2+ or Gd3+ interferes with the phosphate-induced upregulation of NFAT5, SGK1, ORAI1,2,3, STIM1,2 and SOCE in megakaryocytes. To this end, human megakaryocytic Meg-01 cells were treated with 2 mM ß-glycerophosphate for 24 h in the absence and presence of either 1.5 mM MgCl2 or 50 µM GdCl3. Transcript levels were estimated utilizing q-RT-PCR, protein abundance by Western blotting, cytosolic Ca2+ concentration ([Ca2+]i) by Fura-2 fluorescence and SOCE from the increase in [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). As a result, Mg2+ and Gd3+ upregulated CaSR and blunted or virtually abolished the phosphate-induced upregulation of NFAT5, SGK1, ORAI1,2,3, STIM1,2 and SOCE in megakaryocytes. In conclusion, Mg2+ and the CaSR agonist Gd3+ interfere with phosphate-induced dysregulation of [Ca2+]i in megakaryocytes.


Assuntos
Sinalização do Cálcio , Gadolínio/farmacologia , Cloreto de Magnésio/farmacologia , Megacariócitos/efeitos dos fármacos , Proteína ORAI1/metabolismo , Células Cultivadas , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Megacariócitos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Theranostics ; 11(10): 4655-4671, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754019

RESUMO

Rationale: Aurora kinase A (Aurora-A), which is required for mitosis, is a therapeutic target in various tumors. Targeting Aurora-A led to an increase in the differentiation and polyploidization of megakaryocytes both in vivo and in vitro. However, the mechanisms involved in controlling megakaryocyte differentiation have not been fully elucidated. Methods: Conditional Aurka knockout mice were generated. B cell development, platelet development and function were subsequently examined. Proplatelet formation, in vivo response to mTPO, post-transfusion experiment, colony assay, immunofluorescence staining and quantification, and ChIP assay were conducted to gain insights into the mechanisms of Aurka loss in megakaryocytopoiesis. Results: Loss of Aurka in CD19+ B cells impaired B cell development in association with an increase in the number of platelets in peripheral blood (PB). Surprisingly, thrombopoietin (TPO) production and IL-6 were elevated in the plasma in parallel with an increase in the number of differentiated megakaryocytes in the bone marrow (BM) of Aurkaf/f;Cd19Cre/+ mice. Interestingly, compared with that of the Aurkaf/f mice, a higher number of CD19+ B cells close to megakaryocytes was observed in the BM of the Aurkaf/f;Cd19Cre/+ mice. Moreover, Aurka loss in CD19+ B cells induced signal transducer and activator of transcription-3 (STAT3) activation. Inhibition of STAT3 reduced the Tpo mRNA levels. ChIP assays revealed that STAT3 bound to the TPO promoter. Additionally, STAT3-mediated TPO transcription was an autocrine effect provoked by IL-6, at least partially. Conclusions: Deletion of Aurka in CD19+ B cells led to an increase in IL-6 production, promoting STAT3 activation, which in turn contributed to TPO transcription and megakaryocytopoiesis.


Assuntos
Aurora Quinase A/genética , Linfócitos B/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Trombopoese/genética , Trombopoetina/metabolismo , Animais , Antígenos CD19/metabolismo , Tempo de Sangramento , Hepatócitos/metabolismo , Volume Plaquetário Médio , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Contagem de Plaquetas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombopoetina/farmacologia
9.
RNA Biol ; 18(11): 1953-1968, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33586614

RESUMO

Specific miRNA in immune thrombocytopenia (ITP) was screened to explore its intervention effects and mechanisms in ITP. MTT assay and CFSE staining were used to detect the effects of gradient concentrations of thrombopoietin (TPO) on cell proliferation. Expressions of differentially expressed miRNAs were analysed via qRT-PCR in TPO-induced megakaryocytes and ITP plasma. Effects of miR-557 on cell physiological functions were examined by MTT and flow cytometry. Expressions of miR-557, apoptosis-associated genes and Akt/ERK pathways were detected by qRT-PCR and Western blot as needed. Multinucleation of TPO-induced megakaryocytes was determined by megakaryocyte colonies. The toe skin and intestinal bleeding of the ITP rat model were observed and evaluated. Effects of miR-557 on the numbers of platelets, megakaryocytes, and peripheral blood platelets and the expressions of CD4+ T cells, Treg cells, TGF-ß, IL-6 and miR-557 in the ITP rats were detected by Giemsa staining, flow cytometry, ELISA and qRT-PCR. MiR-557 was identified as an specific miRNA associated with both ITP and TPO treatment. MiR-557 inhibitor enhanced the physiological functions of TPO-induced megakaryocytes, while miR-557 mimic had the opposite effect. At the molecular level, the expressions of miR-557, cleaved Caspase-3 and Bax were further silenced by inhibitor, on the contrary, the expressions of bcl-2, p-Akt and p-ERK were upregulated. Animal experiments showed that, miR-557 inhibitor increased the numbers of platelets and megakaryocytes, and improved the symptoms of ITP model rats. Our results indicated that miR-557 inhibitor improved ITP by regulating apoptosis-related genes and cellular immunity and activating the Akt/ERK pathway.


Assuntos
Plaquetas/patologia , Diferenciação Celular , Sistema de Sinalização das MAP Quinases , Megacariócitos/patologia , MicroRNAs/genética , Púrpura Trombocitopênica Idiopática/patologia , Animais , Apoptose , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Estudos de Casos e Controles , Proliferação de Células , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Púrpura Trombocitopênica Idiopática/genética , Púrpura Trombocitopênica Idiopática/metabolismo , Ratos , Trombopoetina/administração & dosagem
10.
Cell Signal ; 80: 109924, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33444776

RESUMO

Toll-like receptors (TLRs) are a class of proteins (patterns recognition receptors-PRRs) capable of recognizing molecules frequently found in pathogens (that are so-called pathogen-associated molecular patterns-PAMPs), they play a key role in the initiation of innate immune response by detecting PAMPs. Our findings show that the functional effects of TLRs co-stimulation on megakaryocytopoiesis. A single cell may receive multiple signal inputs and we consider that multiple TLRs are likely triggered during infection by multiple PAMPs that, in turn, might be involved in infection driven megakaryocytopoiesis, and the present study provide the evidence for the megakaryocytic effects of TLRs co-stimulation.


Assuntos
NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Linhagem Celular Tumoral , Humanos , Integrina beta3/genética , Integrina beta3/metabolismo , Lipopolissacarídeos/farmacologia , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/genética , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Receptor 2 Toll-Like/química , Receptor 4 Toll-Like/química , Zimosan/farmacologia
11.
Toxicol Ind Health ; 37(3): 164-172, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33506746

RESUMO

Air pollution is a worldwide public health issue and it is associated with millions of premature deaths due to cancer, thrombosis, and pulmonary and cardiovascular diseases. Thrombosis is the excessive clotting that blocks a blood vessel, and its etiology is multifactorial. In recent years, growing evidence has linked air pollution, especially particulate matter (PM) and metals, to the development of thrombosis. PM and metals induce lung and systemic inflammation and oxidative stress that are frequent mechanisms in thrombosis. Platelets are important effectors of physiological hemostasis and pathological thrombosis. They are responsible for the formation of the initial plug and are important in the cellular model of coagulation. Therefore, any changes in their morphology or function or an increase in activation could be extremely relevant in thrombosis. Megakaryocytes (MKs) in the bone marrow and in the lungs are the precursor cells of platelets, and the latter is the first organ injured by air pollution. There is substantial evidence of the effect that PM and metals have on platelets, but there is almost no research about the effect of PM and metals on MKs. It is very likely that the alterations produced by air pollution originate in these cells. In this article, we review the biology of MKs and platelets and their role in particulate air pollution-related thrombosis to emphasize the need for further research in this field.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Plaquetas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Material Particulado/efeitos adversos , Trombose/etiologia , Plaquetas/metabolismo , Humanos , Trombose/induzido quimicamente
12.
Thromb Haemost ; 121(2): 192-205, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32961571

RESUMO

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.


Assuntos
Adenina/análogos & derivados , Plaquetas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Trombopoese/efeitos dos fármacos , Adenina/farmacologia , Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Animais , Plaquetas/citologia , Linhagem Celular , Humanos , Megacariócitos/citologia , Camundongos Endogâmicos C57BL
13.
J Cell Physiol ; 236(2): 1445-1453, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32696508

RESUMO

Endocannabinoids are well-known regulators of neurotransmission by activating the cannabinoid (CB) receptors. Endocannabinoids are being used extensively for the treatment of various neurological disorders such as Alzheimer's and Parkinson's diseases. Although endocannabinoids are well studied in cell survival, proliferation, and differentiation in various neurological disorders and several cancers, the functional role in the regulation of blood cell development is less examined. In the present study, virodhamine, which is an agonist of CB receptor-2, was used to examine its effect on megakaryocytic development from a megakaryoblastic cell. We observed that virodhamine increases cell adherence, cell size, and cytoplasmic protrusions. Interestingly, we have also observed large nucleus and increased expression of megakaryocytic marker (CD61), which are the typical hallmarks of megakaryocytic differentiation. Furthermore, the increased expression of CB2 receptor was noticed in virodhamine-induced megakaryocytic cells. The effect of virodhamine on megakaryocytic differentiation could be mediated through CB2 receptor. Therefore, we have studied virodhamine induced molecular regulation of megakaryocytic differentiation; mitogen-activated protein kinase (MAPK) activity, mitochondrial function, and reactive oxygen species (ROS) production were majorly affected. The altered mitochondrial functions and ROS production is the crucial event associated with megakaryocytic differentiation and maturation. In the present study, we report that virodhamine induces megakaryocytic differentiation by triggering MAPK signaling and ROS production either through MAPK effects on ROS-generating enzymes or by the target vanilloid receptor 1-mediated regulation of mitochondrial function.


Assuntos
Endocanabinoides/metabolismo , Hematopoese/genética , Receptor CB2 de Canabinoide/genética , Canais de Cátion TRPV/genética , Ácidos Araquidônicos/metabolismo , Canabinoides/farmacologia , Adesão Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Endocanabinoides/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematopoese/efeitos dos fármacos , Humanos , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor CB1 de Canabinoide
14.
Methods Mol Biol ; 2180: 539-554, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32797433

RESUMO

Current research in the field of transfusion medicine is focused on developing innovative approaches to generate populations of functional megakaryocytes (MKs) ex vivo. This may open perspectives to establish alternative therapies for donor platelet transfusion in the management of thrombocytopenic patients and pave the way for novel regenerative approaches. Efficient cryopreservation techniques can provide the opportunity for long-term storage and accumulation of necessary amounts of MKs in a ready-to-use manner. However, in this case, besides the viability, it is crucial to consider the recovery of functional MK properties after the impact of freezing. In this chapter, the possibility to cryopreserve iPSC-derived MKs is described. In particular, the methods for a comprehensive analysis of phenotypic and functional features of MKs after cryopreservation are proposed. The use of cryopreserved in vitro-produced MKs may benefit to the field of transfusion medicine to overcome the lack of sufficient blood donors.


Assuntos
Plaquetas/citologia , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Criopreservação/métodos , Crioprotetores/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Megacariócitos/citologia , Animais , Plaquetas/efeitos dos fármacos , Diferenciação Celular , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos
15.
Mol Med Rep ; 23(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33179101

RESUMO

Huangqi, the dried root of Radix Astragali, is an essential herb in Traditional Chinese Medicine and has been used to promote hematopoiesis for centuries. Astragalus polysaccharide (ASPS), the bioactive compound of Huangqi, serves a crucial role in hematopoiesis. The aim of the present study was to investigate the hematopoietic effects, in particular the thrombopoietic effects, and the molecular mechanisms of ASPS using an irradiation­induced myelosuppressive mouse model. Colony­forming unit assays, flow cytometric analysis of apoptosis, ELISAs, Giemsa staining and western blotting were performed to determine the hematopoietic and anti­apoptotic effects of ASPS. The results demonstrated that ASPS enhanced the recovery of red blood cells at day 21 following treatment, as well as platelets and white blood cells at day 14. In addition, ASPS promoted colony formation in all lineages (megakaryocytes, granulocyte monocytes, erythroid cells and fibroblasts). The morphological study of the bone marrow demonstrated that tri­lineage hematopoiesis was preserved in the ASPS­ and thrombopoietin (TPO)­treated groups compared with the control group. The overall cellularity (mean total cell count/area) of the ASPS­treated group was similar to that of the TPO­treated group. Additionally, in vitro experiments indicated that treatment with 100 µg/ml ASPS exhibited the maximum effect on colony formation. ASPS attenuated cell apoptosis in megakaryocytic cells via inhibiting the mitochondrial caspase­3 signaling pathway. In conclusion, ASPS promoted hematopoiesis in irradiated myelosuppressive mice possibly via enhancing hematopoietic stem/progenitor cell proliferation and inhibiting megakaryocytes apoptosis.


Assuntos
Medicamentos de Ervas Chinesas/química , Megacariócitos/citologia , Polissacarídeos/administração & dosagem , Lesões Experimentais por Radiação/tratamento farmacológico , Trombocitopenia/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Astragalus propinquus , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Injeções Intraperitoneais , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/efeitos da radiação , Camundongos , Polissacarídeos/farmacologia , Lesões Experimentais por Radiação/complicações , Lesões Experimentais por Radiação/metabolismo , Trombocitopenia/etiologia
16.
Exp Hematol ; 91: 22-31, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32976949

RESUMO

The discovery that the immunomodulatory imide drugs (IMiDs) possess antitumor properties revolutionized the treatment of specific types of hematological cancers. Since then, much progress has been made in understanding why the IMiDs are so efficient in targeting the malignant clones in difficult-to-treat diseases. Despite their efficacy, IMiD resistance arises eventually. Herein we summarize the mechanisms of sensitivity and resistance to lenalidomide in del(5q) myelodysplastic syndrome and multiple myeloma, two diseases in which these drugs are at the therapeutic frontline. Understanding the molecular and cellular mechanisms underlying IMiD efficacy and resistance may allow development of specific strategies to eliminate the malignant clone in otherwise incurable diseases.


Assuntos
Antineoplásicos/farmacologia , Fatores Imunológicos/farmacologia , Lenalidomida/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Anemia Macrocítica/tratamento farmacológico , Anemia Macrocítica/fisiopatologia , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Deleção Cromossômica , Cromossomos Humanos Par 5 , Citocinas/metabolismo , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fator de Transcrição Ikaros/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Lenalidomida/uso terapêutico , Megacariócitos/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/fisiopatologia , Proteínas de Neoplasias/antagonistas & inibidores , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/fisiopatologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/fisiologia
17.
Biosci Rep ; 40(8)2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32779719

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Plaquetas/metabolismo , Megacariócitos/metabolismo , Trombopoese , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose , Plaquetas/efeitos dos fármacos , Linhagem Celular Tumoral , Dobutamina/farmacologia , Regulação da Expressão Gênica , Humanos , Megacariócitos/efeitos dos fármacos , Transdução de Sinais , Trombopoese/efeitos dos fármacos , Transativadores/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Verteporfina/farmacologia , Proteínas de Sinalização YAP
18.
Aging (Albany NY) ; 12(13): 13633-13646, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32651992

RESUMO

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.


Assuntos
Megacariócitos/efeitos dos fármacos , Melatonina/farmacologia , Lesões Experimentais por Radiação/prevenção & controle , Trombocitopenia/prevenção & controle , Trombopoese/efeitos dos fármacos , Animais , Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Plaquetas/efeitos da radiação , Medula Óssea/efeitos dos fármacos , Medula Óssea/fisiologia , Medula Óssea/efeitos da radiação , Caspases/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/efeitos adversos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Masculino , Megacariócitos/fisiologia , Melatonina/uso terapêutico , Camundongos , Mitocôndrias/metabolismo , Neoplasias/terapia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Lesões Experimentais por Radiação/sangue , Lesões Experimentais por Radiação/etiologia , Receptores de Melatonina/antagonistas & inibidores , Receptores de Melatonina/metabolismo , Células-Tronco/efeitos dos fármacos , Trombocitopenia/sangue , Trombocitopenia/etiologia , Trombopoese/efeitos da radiação , Irradiação Corporal Total
19.
Blood ; 136(21): 2410-2415, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32599615

RESUMO

Although cytokine-mediated expansion of human hematopoietic stem cells (HSCs) can result in high yields of hematopoietic progenitor cells, this generally occurs at the expense of reduced bone marrow HSC repopulating ability, thereby limiting potential therapeutic applications. Because bromodomain-containing proteins (BCPs) have been demonstrated to regulate mouse HSC self-renewal and stemness, we screened small molecules targeting various BCPs as potential agents for ex vivo expansion of human HSCs. Of 10 compounds tested, only the bromodomain and extra-terminal motif inhibitor CPI203 enhanced the expansion of human cord blood HSCs without losing cell viability in vitro. The expanded cells also demonstrated improved engraftment and repopulation in serial transplantation assays. Transcriptomic and functional studies showed that the expansion of long-term repopulating HSCs was accompanied by synchronized expansion and maturation of megakaryocytes consistent with CPI203-mediated reprogramming of cord blood hematopoietic stem and progenitor cells. This approach may therefore prove beneficial for ex vivo gene editing, for enhanced platelet production, and for the improved usage of cord blood for transplantation research and therapy.


Assuntos
Acetamidas/farmacologia , Azepinas/farmacologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Reprogramação Celular/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Transcriptoma/efeitos dos fármacos
20.
Biomed Pharmacother ; 129: 110407, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32570117

RESUMO

PURPOSE: Human epidermal growth factor receptor 2 (HER2) is overexpressed in 15-20% of patients with breast cancer. HER2 overexpression is the result of a genetic alteration and this marker is associated with poor clinical outcomes. HER2-targeted therapy can significantly improve the prognosis of patients with either early or advanced HER2-positive breast cancer. One such therapy is the antibody drug conjugate (ADC) trastuzumab emtansine (T-DM1), a combination of trastuzumab and the cytotoxic antimicrotubule agent DM1. After T-DM1 binds HER2, DM1 is subsequently released into the cell. T-DM1 is generally well tolerated and has a relatively low incidence of adverse events. However, there are clinical concerns regarding T-DM1-induced high-grade thrombocytopenia. METHODS: Here, we summarize the incidence of thrombocytopenia from several clinical trials and review experimental studies to explore the causes for T-DM1-induced thrombocytopenia. Progress in several other ADCs targeting HER2-positive breast cancer was also reviewed. CONCLUSIONS: We conclude that T-DM1 uptake by megakaryocytes occurs through either Fcγ receptor binding or through pinocytosis, and we suggest several methods through which these processes could be interrupted to potentially improve the clinical safety of T-DM1. More generally, we recommend that toxicity should be carefully addressed during the development of ADCs.


Assuntos
Ado-Trastuzumab Emtansina/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/efeitos adversos , Megacariócitos/efeitos dos fármacos , Trombocitopenia/induzido quimicamente , Ado-Trastuzumab Emtansina/sangue , Animais , Antineoplásicos Imunológicos/sangue , Feminino , Humanos , Imunoconjugados/sangue , Incidência , Megacariócitos/metabolismo , Pinocitose , Receptores de IgG/sangue , Trombocitopenia/sangue , Trombocitopenia/diagnóstico , Trombocitopenia/epidemiologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA