Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Phytomedicine ; 110: 154637, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36610353

RESUMEN

BACKGROUND: Non-peptide thrombopoietin receptor (TPOR) agonists are promising therapies for the mitigation and treatment of thrombocytopenia. However, only few agents are available as safe and effective for stimulating platelet production for thrombocytopenic patients in the clinic. PURPOSE: This study aimed to develop a novel small molecule TPOR agonist and investigate its underlying regulation of function in megakaryocytes (MKs) differentiation and thrombopoiesis. METHODS: A potential active compound that promotes MKs differentiation and thrombopoiesis was obtained by machine learning (ML). Meanwhile, the effect was verified in zebrafish model, HEL and Meg-01 cells. Next, the key regulatory target was identified by Drug Affinity Responsive Target Stabilization Assay (DARTS), Cellular Thermal Shift Assay (CETSA), and molecular simulation experiments. After that, RNA-sequencing (RNA-seq) was used to further confirm the associated pathways and evaluate the gene expression induced during MK differentiation. In vivo, irradiation (IR) mice, C57BL/6N-TPORem1cyagen (Tpor-/-) mice were constructed by CRISPR/Cas9 technology to examine the therapeutic effect of TMEA on thrombocytopenia. RESULTS: A natural chemical-structure small molecule TMEA was predicted to be a potential active compound based on ML. Obvious phenotypes of MKs differentiation were observed by TMEA induction in zebrafish model and TMEA could increase co-expression of CD41/CD42b, DNA content, and promote polyploidization and maturation of MKs in HEL and Meg-01 cells. Mechanically, TMEA could bind with TPOR protein and further regulate the PI3K/AKT/mTOR/P70S6K and MEK/ERK signal pathways. In vivo, TMEA evidently promoted platelet regeneration in mice with radiation-induced thrombocytopenia but had no effect on Tpor-/- and C57BL/6 (WT) mice. CONCLUSION: TMEA could serve as a novel TPOR agonist to promote MKs differentiation and thrombopoiesis via mTOR and ERK signaling and could potentially be created as a promising new drug to treat thrombocytopenia.


Asunto(s)
Trombocitopenia , Trombopoyesis , Animales , Ratones , Diferenciación Celular , Megacariocitos , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Pez Cebra/metabolismo , Sistema de Señalización de MAP Quinasas , Receptores de Trombopoyetina/antagonistas & inhibidores
2.
Int J Mol Sci ; 23(22)2022 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-36430539

RESUMEN

BACKGROUND: Cibotii rhizoma (CR) is a famous traditional Chinese medicine (TCM) used to treat bleeding, rheumatism, lumbago, etc. However, its therapeutic effects and mechanism against thrombocytopenia are still unknown so far. In the study, we investigated the effects of aqueous extracts of Cibotii rhizoma (AECRs) against thrombocytopenia and its molecular mechanism. METHODS: Giemsa staining, phalloidin staining, and flow cytometry were performed to measure the effect of AECRs on the megakaryocyte differentiation in K562 and Meg-01 cells. A radiation-induced thrombocytopenia mouse model was constructed to assess the therapeutic actions of AECRs on thrombocytopenia. Network pharmacology and experimental verification were carried out to clarify its mechanism against thrombocytopenia. RESULTS: AECRs promoted megakaryocyte differentiation in K562 and Meg-01 cells and accelerated platelet recovery and megakaryopoiesis with no systemic toxicity in radiation-induced thrombocytopenia mice. The PI3K/AKT, MEK/ERK, and JAK2/STAT3 signaling pathways contributed to AECR-induced megakaryocyte differentiation. The suppression of the above signaling pathways by their inhibitors blocked AERC-induced megakaryocyte differentiation. CONCLUSIONS: AECRs can promote megakaryopoiesis and thrombopoiesis through activating PI3K/AKT, MEK/ERK, and JAK2/STAT3 signaling pathways, which has the potential to treat radiation-induced thrombocytopenia in the clinic.


Asunto(s)
Trombocitopenia , Trombopoyesis , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo
3.
Phytomedicine ; 102: 154150, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35569185

RESUMEN

BACKGROUND: Thrombocytopenia (TP) remains a challenge in clinical hematology. TP may have serious consequences, such as recurrent skin and mucosal bleeding and increased risk of intracranial and internal organ hemorrhage. However, effective and safe therapeutic drugs for the long-term management of TP are still lacking. PURPOSE: This study aimed to identify more effective active compounds for TP therapy. METHODS: Liquid chromatography-mass spectrometry-nuclear magnetic resonance analysis was used to confirm the medicinal species and chemical structure of Hirsutine (HS). The proliferation of HS was examined by Cell Counting Kit (CCK-8) assay on cells lines. The effect of HS on megakaryocyte differentiation was analyzed by evaluating the expression of CD41, CD42b, and DNA ploidy via flow cytometry (FCM). The morphology of megakaryocytes and intermediate cells was observed using an optical microscope. K562 cells were then stained with Giemsa and benzidine. qRT-PCR was used to examine the mRNA expression of GATA-1, GATA-2, FOG-1, TAL-1, RUNX-1, NF-E2, and KLF-1 in K562 cells. Protein levels of the transcription factors were analyzed by western blotting. An MEK inhibitor was used to verify the relationship between the MEK/ERK signaling pathway and CD41/CD42b (FCM), FOG-1, and TAL-1. The Kunming thrombocytopenia mouse model was established by X-ray irradiation (4 Gy) and used to test HS activity and related hematopoietic organ index in vivo. Finally, computer simulations of molecular docking were used to predict the binding energies between HS-MEK and HS-ERK. RESULTS: We preliminarily identified HS by screening a plant-sourced compound library for natural compounds with megakaryocytic differentiation and maturation (MKD/MKM)-promoting activity. We found that HS not only enhanced MKD/MKM of K562 and Meg01 cells, but also suppressed the decline of peripheral platelet levels in X-ray-induced myelosuppressive mice. In addition, HS promoted MKD via activation of MEK-ERK-FOG1/TAL1 signaling, which may be the key molecular mechanism of HS action in TP treatment. Molecular docking simulations further verified that HS targets the signaling protein MEK with high-affinity. CONCLUSION: In this study, we report for the first time that hirsutine boosts MKD/MKM through the MEK/ERK/FOG1/TAL1 signaling pathway and thus represents a promising treatment option for TP.


Asunto(s)
Trombocitopenia , Trombopoyesis , Alcaloides , Animales , Diferenciación Celular , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Transducción de Señal , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Factores de Transcripción/metabolismo
4.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153091

RESUMEN

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.


Asunto(s)
16,16-Dimetilprostaglandina E2/uso terapéutico , Síndrome de Radiación Aguda/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Plaquetas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Trastornos Hemorrágicos/tratamiento farmacológico , Lisinopril/uso terapéutico , Megacariocitos/efectos de los fármacos , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Síndrome de Radiación Aguda/complicaciones , Animales , Plaquetas/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Proteína C-Reactiva/análisis , Radioisótopos de Cesio , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de la radiación , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/efectos de la radiación , Femenino , Rayos gamma/efectos adversos , Trastornos Hemorrágicos/etiología , Megacariocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Selectina-P/análisis , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de la radiación , Factor Plaquetario 4/análisis , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/etiología , Trombocitopenia/etiología , Trombopoyesis/efectos de la radiación , Irradiación Corporal Total , Factor de von Willebrand/análisis
5.
J Thromb Haemost ; 19(8): 2029-2043, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33501731

RESUMEN

BACKGROUND: Chemotherapy-induced thrombocytopenia (CIT) can increase the risk of bleeding, which may delay or prevent the administration of anticancer treatment schedules. Photobiomodulation therapy (PBMT), a non-invasive physical treatment, has been proposed to improve thrombocytopenia; however, its underlying regulatory mechanism is not fully understood. OBJECTIVE: To further investigate the mechanism of thrombopoietin (TPO) in megakaryocytopoiesis and thrombopoiesis. METHODS: Multiple approaches such as western blotting, cell transfection, flow cytometry, and animal studies were utilized to explore the effect and mechanism of PBMT on thrombopoiesis. RESULTS: PBMT prevented a severe drop in platelet count by increasing platelet production, and then ameliorated CIT. Mechanistically, PBMT significantly upregulated hepatic TPO expression in a thrombocytopenic mouse model, which promoted megakaryocytopoiesis and thrombopoiesis. The levels of TPO mRNA and protein increased by PBMT via the Src/ERK/STAT3 signaling pathway in hepatic cells. Furthermore, the generation of the reactive oxygen species was responsible for PBMT-induced activation of Src and its downstream target effects. CONCLUSIONS: Our research suggests that PBMT is a promising therapeutic strategy for the treatment of CIT.


Asunto(s)
Terapia por Luz de Baja Intensidad , Trombocitopenia , Animales , Plaquetas , Megacariocitos , Ratones , Especies Reactivas de Oxígeno , Transducción de Señal , Trombocitopenia/terapia , Trombopoyesis , Trombopoyetina
6.
Blood Adv ; 2(17): 2262-2272, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30206099

RESUMEN

During maturation, megakaryocytes (MKs) express ß1-tubulin (TUBB1) and rearrange their microtubule components to enlarge, form proplatelets, and eventually release platelets. The development of a platform to identify in vitro conditions that would efficiently promote MK development could potentially enable large-scale platelet production. Here, we show that an immortalized MK cell line (imMKCL) genetically modified to express the ß1-tubulin-Venus reporter provides a practical system to efficiently monitor the in vitro production of platelet-like particles (PLPs). The Venus transgene was inserted downstream of the TUBB1 locus in imMKCLs using CRISPR/Cas9, and the expression was visualized by Venus fluorescence intensity. This imMKCL reporter line was then used for high-throughput drug screening. We identified several compounds that significantly improved the efficiency of PLP production in vitro under feeder-free conditions and showed a significant tendency to recover platelets in vivo in a mouse thrombocytopenia model induced by anti-GPIbα antibody administration. Interestingly, most of these compounds, including a WNT signaling pathway inhibitor, Wnt-C59, antagonized the aryl hydrocarbon receptor (AhR) to increase PLP production, confirming the crucial role of AhR inhibition in MK maturation. Consistently, small interfering RNA treatment against AhR increased the Venus intensity and PLP production. TCS 359, an FLT3 inhibitor, significantly increased PLP production independently of FLT3 or AhR. This study highlights the usefulness of the ß1-tubulin reporter MK line as a useful tool to study the mechanisms underlying thrombopoiesis and to identify novel inducers of ex vivo platelet production.


Asunto(s)
Plaquetas/citología , Descubrimiento de Drogas/métodos , Genes Reporteros/genética , Megacariocitos/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Luciferasas/genética , Masculino , Megacariocitos/citología , Ratones , Ratones Endogámicos C57BL , Receptores de Hidrocarburo de Aril/metabolismo , Trombopoyesis
7.
Biochem Biophys Res Commun ; 505(1): 168-175, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30243726

RESUMEN

Megakaryocytopoiesis results in the formation of platelets, which are essential for hemostasis. Decreased production or increased destruction of platelets can cause thrombocytopenia, in which platelet transfusion is the mode of treatment. The present study is aimed in generation of megakaryocytes (MKs) and platelet from human hematopoietic stem cells (HSCs). The purity of HSCs was assessed through Flow cytometry and immunocytochemistry (ICC) studies. These pure HSCs were induced with thrombopoietin (TPO), similarly with Andrographis paniculata extract (APE) for 21 days to generate MKs. The APE is mainly composed of andrographolide which stimulates TPO from the liver, and this binds to CD110 present on the surface of HSCs and triggers the proliferation of HSCs and initiate higher MKs population subsequently, a large number of platelets. The results of the present study showed increased proliferation of HSCs grown in the presence of APE and revealed a high population of CD41a and CD42b positive MKs as enumerated by Flow cytometry compared with TPO induced MKs. These results also concurred with qRT-PCR and western blot analysis. The scanning electron microscopy (SEM) revealed the morphology of differentiated MKs and platelets were similar to human blood platelets. The differentiated MKs in APE exhibited polyploidy up to 32 N while TPO induced MKs showed polyploidy of 8 N, these results corroborated with colony forming unit assay. On thrombin stimulation, high expression of P-selectin (CD62p) and fibrinogen binding were detected in APE induced platelets. Autologous transplantation of platelets generated from APE may be a useful option in thrombocytopenia condition.


Asunto(s)
Plaquetas/citología , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Megacariocitos/citología , Andrographis paniculata , Células Cultivadas , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Microscopía Electrónica de Rastreo , Extractos Vegetales/farmacología , Trombopoyesis/efectos de los fármacos , Trombopoyesis/genética , Trombopoyetina/farmacología
8.
Clin Lab ; 64(3): 329-332, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29739118

RESUMEN

BACKGROUND: Iron deficiency anemia (IDA) is characterized by depletion of total body iron stores or a poor supply of plasma iron. By contrast, chronic inflammation makes iron unavailable for hematopoiesis through a cytokinemediated cascade and leads to a condition known as anemia of chronic disease (AOC). However, the laboratory data regarding the regulatory role of iron metabolism on platelet count has not been fully discussed yet. In this study, we investigated the relationship between iron status and platelet production according to different anemic mechanisms representing different iron metabolisms. METHODS: The study included a total of 759 specimens. Blood samples were obtained through venipuncture. The complete blood count was measured using an Advia 2120 (Siemens Healthcare Diagnostics Inc., USA). Biochemical indices including iron level were estimated using a Toshiba chemical analyzer (Toshiba, Japan). RESULTS: In the AOC group, we found a significant relationship between platelet count and serum iron level (p < 0.27), whereas there was no correlation in the IDA group. Moreover, when the AOC patient group was subdivided by serum iron level, a remarkable difference was observed as follows. The platelet count was significantly correlated with serum iron level only in the AOC group with decreased serum iron levels (serum iron < 50 µg/dL) (p < 0.0001), while there was no correlation in the AOC group with normal serum iron levels (serum iron 50 - 100 µg/dL). CONCLUSIONS: Iron deficiency in AOC involves upregulated hepcidin production induced by elevated inflammatory cytokines. This can cause increased iron sequestration in macrophages and decreased iron absorption for bone marrow. The condition of decreased megakaryocytic iron supply makes megakaryocytes with higher ploidy which can release more platelets than lower ploidy. Moreover, reactive thrombocytosis in inflammatory states occurs by cytokine cascades involving interleukin 6 and thrombopoietin in AOC. These two features may enhance thrombocytosis in patients of AOC with decreased iron level. In the future, further study should be performed to elucidate regulating mechanism of iron metabolism for megakaryopoiesis in AOC patients, and guide proper supplemental therapy of iron to decrease thrombotic risk due to reactive thrombocytosis in various kinds of anemia.


Asunto(s)
Anemia/sangre , Hierro/sangre , Megacariocitos/metabolismo , Trombopoyesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Ferropénica/sangre , Enfermedad Crónica , Citocinas/sangre , Femenino , Humanos , Hierro/metabolismo , Masculino , Megacariocitos/citología , Persona de Mediana Edad , Recuento de Plaquetas , Adulto Joven
9.
Phytomedicine ; 43: 135-139, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29747746

RESUMEN

BACKGROUND: Hepatoprotective activity along with improved survival percentage and hematological parameters prior to whole body irradiation were reported with Justicia adhatoda extracts. PURPOSE: To evaluate the thrombopoietic potential of Justicia adhatoda L. leaf extract in megakaryocyte differentiation METHODS: Ethanol extracts were prepared using soxhlet extraction method, and IC50 value was determined. The effect of ethanol extracts obtained from Justicia adhatoda on megakaryocyte maturation and development in megakaryocytic Dami cell lines was tested. Expression of megakaryocyte specific markers, CD61 and CD41, were assessed using flow cytometry and fluorescence microscopy. In addition, cell cycle analysis and mitochondrial membrane potential were analyzed by flow cytometry. Gene expression analysis was performed using qRT-PCR. RESULTS: At a concentration of 40 µg/ml, the leaf extracts of Justicia adhatoda for 72 h induced the megakaryocytic features in megakaryocytic Dami cell lines. The megakaryocyte specific markers, CD41 and CD61, were up-regulated (2.2 and 12.4 fold, respectively), and more number of cells entered into synthetic (S) and G2/M phase as compared with untreated cell (23.1% vs 16.6% and 70.2% vs 42.3%, respectively) showing maturation. RUNX1 (a transcription factor essential for embryonic hematopoiesis and adult megkaryocyte maturation) and c-Mpl (the receptor for TPO) were upregulated, and the suppressor of cytokine signaling (SOCS) 1 and SOCS3 were down-regulated upon treatment with Justicia adhatoda. Justicia adhatoda enhanced mitochondrial ROS generation by 28-fold, increased the permeability of mitochondrial membrane and showed an inverse correlation in superoxide dismutase levels. CONCLUSION: Justicia adhatoda could enhance mitochondrial ROS generation and increase the permeability of mitochondrial membrane, thereby inducing megakaryocytic maturation. Our findings suggest thrombopoietic potential of Justicia adhatoda leaf extract on megakaryocyte differentiation.


Asunto(s)
Género Justicia/química , Megacariocitos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Concentración 50 Inhibidora , Integrina beta3/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Mitocondrias/metabolismo , Hojas de la Planta/química , Plantas Medicinales/química , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Trombopoyesis/efectos de los fármacos
10.
Sci Rep ; 8(1): 5075, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29567949

RESUMEN

Thrombocytopenia or chronic depletion of platelets in blood, could create life-threatening conditions in patients who receive aggressive systemic radiation and chemotherapy. Currently there are no approved agents for the rapid treatment of thrombocytopenia. In the present study, we demonstrate that administration of Orientin, a glycosidic flavonoid or dietary administration of Orientin containing Tulsi (Holy Basil) leaves, results in a significant increase in circulating platelets in a clinically relevant mouse model. No noticeable effects were observed on red blood cells, white blood cells or other hematologic parameters in treated animals indicating that Orientin specificity enhances platelet formation. The gene expression and immunophenotyping of bone marrow revealed that Orientin stimulates megakaryopoiesis specific transcriptional program. A significant increase in colony formation in bone marrow cells from Orientin pretreated mice further complemented the effect of Orientin on progenitor cells. The ex-vivo differentiation of irradiated human peripheral blood CD34+ stem cells demonstrated stimulatory effects of Orientin on megakaryocyte erythrocyte progenitors (MEP). The results show that Orientin, a non-toxic readily available natural product can counter platelet imbalances. Thrombocytopenia also develop as a consequence of multiple hematologic malignancies and side effects of treatments. Dietary supplementation of Orientin containing phytochemicals could be effective as countermeasures and viable therapeutics.


Asunto(s)
Plaquetas/efectos de los fármacos , Flavonoides/administración & dosificación , Glucósidos/administración & dosificación , Ocimum sanctum/química , Trombocitopenia/dietoterapia , Animales , Plaquetas/metabolismo , Plaquetas/patología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Flavonoides/química , Regulación de la Expresión Génica/efectos de los fármacos , Glucósidos/química , Humanos , Ratones , Fitoquímicos/química , Fitoquímicos/uso terapéutico , Trombocitopenia/sangre , Trombocitopenia/patología , Trombopoyesis/efectos de los fármacos
11.
J Nat Med ; 72(1): 211-219, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29019067

RESUMEN

We report in this study novel biochemical activities of peanut skin extract (PEXT) on thrombocytopoiesis. Peanut skin, derived from Arachis hypogaea L., is a traditional Chinese medicine that is used to treat chronic hemorrhage. We have shown that oral administration of PEXT increases the peripheral platelet levels in mice. Recently, we reported a liquid culture system that is useful for investigating megakaryocytopoiesis and thrombocytopoiesis from human CD34+ cells. In this liquid culture system, PEXT was shown to enhance the formation of CD41+/DAPI- cells (platelets), but had no effect on the formation of CD41+/DAPI+ cells (megakaryocytes) or on the DNA content. Furthermore, PEXT selectively stimulated proplatelet formation from cultured mature megakaryocytes and phorbol 12-myristate 13 acetate (PMA)-induced formation of platelet-like particles from Meg01 cells. Despite having no influence on the formation of megakaryocyte colony forming units (CFUs), PEXT increased the size of megakaryocytes during their development from CD34+ cells. PEXT showed no effect on the GATA-1 and NF-E2 mRNA levels, which are known to play an important role in thrombocytopoiesis and, based on the results of a pMARE-Luc (pGL3-MARE-luciferase) assay, had no influence on NF-E2 activation in Meg01 cells. These results suggest that PEXT accelerates proplatelet formation from megakaryocytes but does not influence the development of hematopoietic stem cells into megakaryocytes.


Asunto(s)
Arachis/química , Plaquetas/metabolismo , Megacariocitos/metabolismo , Trombopoyesis/efectos de los fármacos , Animales , Diferenciación Celular , Humanos , Masculino , Ratones
12.
J Nutr Biochem ; 47: 94-105, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28570944

RESUMEN

Hematopoietic stem cells play the vital role of maintaining appropriate levels of cells in blood. Therefore, regulation of their fate is essential for their effective therapeutic use. Here we report the role of polyunsaturated fatty acids (PUFAs) in regulating hematopoiesis which has not been explored well so far. Mice were fed daily for 10 days with n-6/n-3 PUFAs, viz. linoleic acid (LA), arachidonic acid (AA), alpha-linolenic acid and docosahexanoic acid (DHA) in four separate test groups with phosphate-buffered saline fed mice as control set. The bone marrow cells of PUFA-fed mice showed a significantly higher hematopoiesis as assessed using side population, Lin-Sca-1+ckit+, colony-forming unit (CFU), long-term culture, CFU-spleen assay and engraftment potential as compared to the control set. Thrombopoiesis was also stimulated in PUFA-fed mice. A combination of DHA and AA was found to be more effective than when either was fed individually. Higher incorporation of PUFAs as well as products of their metabolism was observed in the bone marrow cells of PUFA-fed mice. A stimulation of the Wnt, CXCR4 and Notch1 pathways was observed in PUFA-fed mice. The clinical relevance of this study was evident when bone marrow-transplanted recipient mice, which were fed with PUFAs, showed higher engraftment of donor cells, suggesting that the bone marrow microenvironment may also be stimulated by feeding with PUFAs. These data indicate that oral administration of PUFAs in mice stimulates hematopoiesis and thrombopoiesis and could serve as a valuable supplemental therapy in situations of hematopoietic failure.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Suplementos Dietéticos , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Grasos Omega-6/uso terapéutico , Hematopoyesis , Trombopoyesis , Regulación hacia Arriba , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Suplementos Dietéticos/efectos adversos , Ácidos Grasos Omega-3/efectos adversos , Ácidos Grasos Omega-6/efectos adversos , Femenino , Regulación de la Expresión Génica , Supervivencia de Injerto , Hematínicos/uso terapéutico , Ratones Congénicos , Ratones Endogámicos C57BL , Receptor Notch1/agonistas , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores CXCR4/agonistas , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Acondicionamiento Pretrasplante/efectos adversos , Proteínas Wnt/agonistas , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
13.
Sci Rep ; 6: 38238, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27901126

RESUMEN

Immune thrombocytopenia (ITP) is an immune-mediated acquired bleeding disorder characterized by abnormally low platelet counts. We reported here the ability of low-level light treatment (LLLT) to alleviate ITP in mice. The treatment is based on noninvasive whole body illumination 30 min a day for a few consecutive days by near infrared light (830 nm) transmitted by an array of light-emitting diodes (LEDs). LLLT significantly lifted the nadir of platelet counts and restored tail bleeding time when applied to two passive ITP models induced by anti-CD41 antibody. The anti-platelet antibody hindered megakaryocyte differentiation from the progenitors, impaired proplatelet and platelet formation, and induced apoptosis of platelets. These adverse effects of anti-CD41 antibody were all mitigated by LLLT to varying degrees, owing to its ability to enhance mitochondrial biogenesis and activity in megakaryocytes and preserve mitochondrial functions in platelets in the presence of the antibody. The observations argue not only for contribution of mitochondrial stress to the pathology of ITP, but also clinical potentials of LLLT as a safe, simple, and cost-effective modality of ITP.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Megacariocitos/efectos de la radiación , Trombocitopenia/radioterapia , Animales , Apoptosis/inmunología , Apoptosis/efectos de la radiación , Diferenciación Celular/inmunología , Megacariocitos/citología , Megacariocitos/inmunología , Ratones Endogámicos C57BL , Recuento de Plaquetas , Trombocitopenia/inmunología , Trombopoyesis/inmunología , Trombopoyesis/efectos de la radiación
14.
Haematologica ; 101(8): 896-908, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27478199

RESUMEN

Constitutional thrombocytopenias result from platelet production abnormalities of hereditary origin. Long misdiagnosed and poorly studied, knowledge about these rare diseases has increased considerably over the last twenty years due to improved technology for the identification of mutations, as well as an improvement in obtaining megakaryocyte culture from patient hematopoietic stem cells. Simultaneously, the manipulation of mouse genes (transgenesis, total or conditional inactivation, introduction of point mutations, random chemical mutagenesis) have helped to generate disease models that have contributed greatly to deciphering patient clinical and laboratory features. Most of the thrombocytopenias for which the mutated genes have been identified now have a murine model counterpart. This review focuses on the contribution that these mouse models have brought to the understanding of hereditary thrombocytopenias with respect to what was known in humans. Animal models have either i) provided novel information on the molecular and cellular pathways that were missing from the patient studies; ii) improved our understanding of the mechanisms of thrombocytopoiesis; iii) been instrumental in structure-function studies of the mutated gene products; and iv) been an invaluable tool as preclinical models to test new drugs or develop gene therapies. At present, the genetic determinants of thrombocytopenia remain unknown in almost half of all cases. Currently available high-speed sequencing techniques will identify new candidate genes, which will in turn allow the generation of murine models to confirm and further study the abnormal phenotype. In a complementary manner, programs of random mutagenesis in mice should also identify new candidate genes involved in thrombocytopenia.


Asunto(s)
Trombocitopenia/etiología , Trombocitopenia/metabolismo , Animales , Autoantígenos/metabolismo , Síndrome de Bernard-Soulier/etiología , Síndrome de Bernard-Soulier/metabolismo , Plaquetas/metabolismo , Diferenciación Celular/genética , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Yoduro Peroxidasa/metabolismo , Proteínas de Unión a Hierro/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Trombocitopenia/diagnóstico , Trombopoyesis , Factores de Transcripción/metabolismo , Síndrome de Wiskott-Aldrich/etiología , Síndrome de Wiskott-Aldrich/metabolismo
15.
Sci Transl Med ; 8(349): 349ra101, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27464749

RESUMEN

Thrombocytopenia is a common hematologic disorder that is managed primarily by platelet transfusions. We report here that noninvasive whole-body illumination with a special near-infrared laser cures acute thrombocytopenia triggered by γ-irradiation within 2 weeks in mice, as opposed to a 5-week recovery time required in controls. The low-level laser (LLL) also greatly accelerated platelet regeneration in the presence of anti-CD41 antibody that binds and depletes platelets, and prevented a severe drop in platelet count caused by a common chemotherapeutic drug. Mechanistically, LLL stimulated mitochondrial biogenesis specifically in megakaryocytes owing to polyploidy of the cells. LLL also protected megakaryocytes from mitochondrial injury and apoptosis under stress. The multifaceted effects of LLL on mitochondria bolstered megakaryocyte maturation; facilitated elongation, branching, and formation of proplatelets; and doubled the number of platelets generated from individual megakaryocytes in mice. LLL-mediated platelet biogenesis depended on megakaryopoiesis and was inversely correlated with platelet counts, which kept platelet biogenesis in check and effectively averted thrombosis even after repeated uses, in sharp contrast to all current agents that stimulate the differentiation of megakaryocyte progenitors from hematopoietic stem cells independently of platelet counts. This safe, drug-free, donor-independent modality represents a paradigm shift in the prophylaxis and treatment of thrombocytopenia.


Asunto(s)
Terapia por Luz de Baja Intensidad/métodos , Trombocitopenia/terapia , Animales , Antígenos CD34/metabolismo , Plaquetas/efectos de la radiación , Células Cultivadas , Citometría de Flujo , Humanos , Megacariocitos/metabolismo , Megacariocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Trombopoyesis/efectos de la radiación
16.
Nat Commun ; 7: 11097, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-27020697

RESUMEN

Mg(2+) plays a vital role in platelet function, but despite implications for life-threatening conditions such as stroke or myocardial infarction, the mechanisms controlling [Mg(2+)]i in megakaryocytes (MKs) and platelets are largely unknown. Transient receptor potential melastatin-like 7 channel (TRPM7) is a ubiquitous, constitutively active cation channel with a cytosolic α-kinase domain that is critical for embryonic development and cell survival. Here we report that impaired channel function of TRPM7 in MKs causes macrothrombocytopenia in mice (Trpm7(fl/fl-Pf4Cre)) and likely in several members of a human pedigree that, in addition, suffer from atrial fibrillation. The defect in platelet biogenesis is mainly caused by cytoskeletal alterations resulting in impaired proplatelet formation by Trpm7(fl/fl-Pf4Cre) MKs, which is rescued by Mg(2+) supplementation or chemical inhibition of non-muscle myosin IIA heavy chain activity. Collectively, our findings reveal that TRPM7 dysfunction may cause macrothrombocytopenia in humans and mice.


Asunto(s)
Citoesqueleto/metabolismo , Homeostasis , Magnesio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPM/metabolismo , Trombopoyesis , Animales , Plaquetas/metabolismo , Humanos , Megacariocitos/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Canales Catiónicos TRPM/deficiencia , Trombocitopenia/metabolismo , Trombocitopenia/patología
17.
Int J Toxicol ; 32(2): 100-12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23559642

RESUMEN

ALXN4100TPO, a thrombopoietin (TPO) receptor agonist, increases platelets, abrogates radiation-induced thrombocytopenia and affords significant survival benefit to lethally irradiated mice. This preliminary nonclinical safety study assessed effects of a single subcutaneous (sc) administration of ALXN4100TPO in CD2F1 mice randomized into naïve, control antibody (ALXN4200, 100 mg/kg), low (1 mg/kg), medium (10 mg/kg), or high (100 mg/kg) ALXN4100TPO doses. End points included clinical observations, body weight changes, hematology, histopathology, pharmacokinetics, pharmacodynamics by measuring platelet counts, and endogenous TPO (eTPO) levels. Salient findings were prominent increase in platelet counts and end cells of myeloid and lymphoid lineages; elevated megakaryopoiesis in bone marrow; and extramedullary hematopoiesis in spleen and liver. Serum ALXN4100TPO levels were maximum 24 hours after administration, with a half-life of 13 days. Endogenous TPO levels were elevated in 10 and 100 mg/kg ALXN4100TPO-treated groups. In conclusion, ALXN4100TPO (1-100 mg/kg, sc) treatment in CD2F1 mice resulted in profound pharmacological changes in the hematopoietic tissue; however, no life-threatening adverse events were observed.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/toxicidad , Receptores de Trombopoyetina/agonistas , Trombopoyetina/farmacocinética , Trombopoyetina/toxicidad , Animales , Anticuerpos Monoclonales Humanizados , Disponibilidad Biológica , Plaquetas/citología , Plaquetas/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratones , Ratones Endogámicos , Recuento de Plaquetas , Bazo/efectos de los fármacos , Bazo/patología , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/patología , Trombopoyesis/efectos de los fármacos
18.
Radiat Res ; 176(6): 716-24, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22026586

RESUMEN

Hematopoietic processes, especially megakaryocytopoiesis and thrombopoiesis, are highly sensitive to extracellular oxidative stresses such as ionizing radiation and chemotherapeutic agents. This study examined the terminal maturation of megakaryocytes and platelet production in hematopoietic stem/progenitor cells (HSPCs) exposed to ionizing radiation. Highly purified CD34(+) cells derived from human placental/umbilical cord blood were exposed to X rays (2 Gy, 150 kVp, 20 mA; 0.5-mm aluminum and 0.3-mm copper filters) at a dose rate of approximately 1 Gy/min and then cultured in a serum-free medium supplemented with thrombopoietin and interleukin-3. The number of cells generated from X-irradiated CD34(+) cells decreased with the time in culture. However, the fraction of CD34(+)Tie-2(+) and CD41(+)Tie-2(+) cells among the total cells generated from X-irradiated cells increased significantly in comparison to nonirradiated controls on day 7. In addition, the CD42a(+) particles, which appeared to be platelets, generated from the X-irradiated HSPCs appeared to be normal. Quantitative real-time reverse transcriptase-polymerase chain reaction analysis of the expression of various genes in cells harvested from the cultures showed that the early hematopoiesis-related genes FLI1, HOXB4 and Tie-2, the cytokine receptor genes KIT and IL3RA, and the oxidative stress-related genes HO1 and NQO1 were upregulated on day 7. These results suggest that normal terminal maturation of megakaryocytes and platelet production occur in residual HSPCs after exposure to ionizing radiation despite the adverse effect of radiation on proliferation and differentiation of HSPCs. Ionizing radiation may have the potential to promote both megakaryocytopoiesis and thrombopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Trombopoyesis/efectos de la radiación , Antígenos CD34/metabolismo , Plaquetas/citología , Plaquetas/efectos de la radiación , Femenino , Sangre Fetal/citología , Regulación de la Expresión Génica/efectos de la radiación , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-3/metabolismo , Megacariocitos/citología , Megacariocitos/efectos de la radiación , Placenta/citología , Placenta/efectos de la radiación , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Dosis de Radiación , Trombopoyetina/metabolismo
19.
BMC Complement Altern Med ; 10: 79, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21176128

RESUMEN

BACKGROUND: Dozens of Traditional Chinese Medicine (TCM) formulas have been used for promotion of "blood production" for centuries, and we are interested in developing novel thrombopoietic medicines from these TCMs. Our previous studies have demonstrated the hematopoietic effects of DangGui BuXue Tong (DBT), a formula composed of Radix Angelicae Sinensis and Radix Astragali in animal and cellular models. As a step further to identify and characterize the active chemical components of DBT, we tested the hematopoietic and particularly, thrombopoietic effects of polysaccharide-enriched fractions from the root of Radix Angelicae Sinensis (APS) in this study. METHODS: A myelosuppression mouse model was treated with APS (10 mg/kg/day). Peripheral blood cells from APS, thrombopoietin and vehicle-treated samples were then counted at different time-points. Using the colony-forming unit (CFU) assays, we determined the effects of APS on the proliferation and differentiation of hematopoietic stem/progenitor cells and megakaryocytic lineages. Using a megakaryocytic cell line M-07e as model, we analyzed the cellular apoptosis progression with and without APS treatment by Annexin V, Mitochondrial Membrane Potential and Caspase 3 assays. Last, the anti-apoptotic effect of APS on cells treated with Ly294002, a Phosphatidylinositol 3-Kinse inhibitor (PI3K) was also tested. RESULTS: In animal models, APS significantly enhanced not only the recovery of platelets, other blood cells and their progenitor cells, but also the formation of Colony Forming Unit (CFU). In M-07e cells, we observed the anti-apoptotic effect of APS. Treatment by Ly294002 alone increased the percentage of cells undergoing apoptosis. However, addition of APS to Ly294002-treated cells significantly reduced the percentage of cells undergoing apoptosis. CONCLUSIONS: APS promotes hematopoiesis and thrombopoiesis in the mouse model. This effect likely resulted from the anti-apoptosis activity of APS and is likely to involve the PI3K/AKT pathway.


Asunto(s)
Angelica sinensis/química , Medicamentos Herbarios Chinos/farmacología , Hematopoyesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Polisacáridos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombopoyesis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Sanguíneas/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Modelos Animales de Enfermedad , Megacariocitos/efectos de los fármacos , Ratones , Morfolinas/farmacología , Raíces de Plantas/química , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos
20.
Exp Hematol ; 37(11): 1364-1377.e4, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19744539

RESUMEN

OBJECTIVE: The signaling by thrombopoietin (TPO) via its receptor, c-MPL, plays a crucial role in the maintenance of hematopoietic stem cells (HSCs). Small-molecule c-MPL agonists have recently been shown to be beneficial in the treatment of thrombocytopenia. However, their effects on HSCs have not yet been explored. In this study, we evaluated the effects of NR-101, a novel small-molecule c-MPL agonist, on the ex vivo expansion of human cord blood (hCB) HSCs. MATERIALS AND METHODS: hCB CD34(+) or CD34(+)CD38(-) hematopoietic stem and progenitor cells were cultured for 7 days in the presence of thrombopoietin (TPO) or NR-101, and then subjected to flow cytometric analyses, colony-forming cell assays, and severe combined immunodeficiency-repopulating cell assays. RESULTS: During a 7-day culture of CD34(+) or CD34(+)CD38(-) hematopoietic stem and progenitor cells, NR-101 efficiently increased their numbers, with a greater than twofold increase compared to TPO, although its effect on megakaryocytopoiesis was comparable to that of TPO. Correspondingly, severe combined immunodeficiency-repopulating cells were increased 2.9-fold during a 7-day culture with NR-101 compared to freshly isolated CD34(+) cells, and 2.3-fold compared to that with TPO. Of note, NR-101 persistently activated signal transducer and activator of transcription (STAT) 5 but not signal transducer and activator of transcription 3. Furthermore, NR-101 induced a long-term accumulation of hypoxia-inducible factor-1alpha protein and enhanced activation of its downstream target genes. CONCLUSION: This is the first time that a small-molecule c-MPL agonist has been demonstrated to promote net expansion of HSCs. NR-101 is more efficient in ex vivo expansion of HSCs than TPO. NR-101 could be a useful tool for the therapeutic manipulation of human HSCs.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Receptores de Trombopoyetina/agonistas , Trombopoyesis/efectos de los fármacos , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Línea Celular Tumoral/citología , Línea Celular Tumoral/efectos de los fármacos , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/trasplante , Trasplante de Células Madre de Sangre del Cordón Umbilical , Subunidad beta Común de los Receptores de Citocinas/genética , Subunidad beta Común de los Receptores de Citocinas/fisiología , ADN Complementario/genética , Evaluación Preclínica de Medicamentos , Sangre Fetal/citología , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Receptor de Interleucina-3/genética , Subunidad alfa del Receptor de Interleucina-3/fisiología , Leucemia Mieloide/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Quimera por Radiación , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/fisiología , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/fisiología , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/efectos de los fármacos , Trombopoyetina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA