Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
Sci Rep ; 7(1): 12184, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28939876

RESUMO

Tumor radiotherapy induces hematopoietic organ damage and reduces thrombocyte counts. Thrombocytopenia is a common disease. Some studies have shown that tRNA synthetase plays not only catalytic tRNA aminoacylation roles, but also functions similarly to cytokines. Recombinant human tyrosyl-tRNA synthetase with a mutated Y341A (rhTyrRS (Y341A)) promotes megakaryocyte migrate from bone marrow to peripheral blood. It would promote megakaryocytes in the lungs adhering to vascular endothelial cells and resulting in the platelet production. The purpose of this research was to investigate the efficacy of rhTyrRS (Y341A) as a therapy for thrombocytopenia and to explore its mechanism of action. We found platelet number was effectively increased by rhTyrRS (Y341A) via platelet count and reticulated platelets (RPs) flow cytometry. We also demonstrated radiation-induced thrombocytopenia could be prevented by rhTyrRS (Y341A). The results of immunohistochemistry and H&E staining showed the number of pulmonary mature megakaryocytes was significantly increased in rhTyrRS (Y341A) treated groups. In transgenic zebrafish larvae, confocal microscopy results showed rhTyrRS (Y341A) promoted the migration and adhesion of megakaryocytes. These results suggested that rhTyrRS (Y341A) promote megakaryocytes in bone marrow migrating to lungs through blood circulation. rhTyrRS (Y341A) may be an effective medicine which could be used to treat patients suffering from thrombocytopenia.


Assuntos
Plaquetas/efeitos dos fármacos , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Tirosina-tRNA Ligase/administração & dosagem , Animais , Animais Geneticamente Modificados , Plaquetas/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neoplasias/radioterapia , Contagem de Plaquetas , Radioterapia/efeitos adversos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Trombocitopenia/etiologia , Trombopoese/efeitos da radiação , Resultado do Tratamento , Tirosina-tRNA Ligase/genética , Peixe-Zebra
4.
Sci Rep ; 6: 38238, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27901126

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Megacariócitos/efeitos da radiação , Trombocitopenia/radioterapia , Animais , Apoptose/imunologia , Apoptose/efeitos da radiação , Diferenciação Celular/imunologia , Megacariócitos/citologia , Megacariócitos/imunologia , Camundongos Endogâmicos C57BL , Contagem de Plaquetas , Trombocitopenia/imunologia , Trombopoese/imunologia , Trombopoese/efeitos da radiação
5.
Sci Transl Med ; 8(349): 349ra101, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27464749

RESUMO

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.


Assuntos
Terapia com Luz de Baixa Intensidade/métodos , Trombocitopenia/terapia , Animais , Antígenos CD34/metabolismo , Plaquetas/efeitos da radiação , Células Cultivadas , Citometria de Fluxo , Humanos , Megacariócitos/metabolismo , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Trombopoese/efeitos da radiação
6.
J Pharmacokinet Pharmacodyn ; 42(6): 709-20, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26341875

RESUMO

A mechanistic model describing the effects of chemotherapy and radiation on platelet counts and endogenous thrombopoietin (eTPO) in mice was developed. Thrombocytopenia was induced in mice by injection of carboplatin followed by the whole body irradiation on days 0, 28, and 56, with platelet and eTPO samples collected over 84 days. The pharmacodynamic model consisted of a series of aging compartments representing proliferating megakaryocyte precursors, megakaryocytes, and platelets with possible eTPO clearance through internalization. The cytotoxic effects of treatment were described by the kinetics of the effect (K-PD) model, and stimulation of platelet production by eTPO was considered to be driven by receptor occupancy. The proposed PD model adequately described the platelet counts and eTPO concentrations in mice by accounting for nadirs and peaks of platelet count, and rebounds in eTPO time course profiles. The estimates of model parameters were in good agreement with their physiological values reported in literature for mice with platelet lifespan of 4.3 days and 185 cMpl receptors per platelet. The predicted duration of the treatment effect was 0.82 h (approximately 5 carboplatin half-lives in mice). The data was not informative about the eTPO stimulatory effect as the nominal precursor production rate was sufficient to account for platelet response to treatment. The model quantified the inverse relationship between eTPO levels and platelet counts and offered an explanation of the tolerance effect observed in the eTPO data. The simulated rebound in free receptors levels correlated with rebounds in eTPO levels. The model suggests that the duration of the toxic effects is determined by the turnover of the proliferating cells in the bone marrow. This indicates that the lifespan of the target cells (megakaryocyte precursors, megakaryocytes and platelets) is a key determinant in the duration of both drug exposure and toxicity due to treatment. The model can be extended to account for pharmacokinetics of exogenous drugs and be applied to analysis of human data.


Assuntos
Antineoplásicos/toxicidade , Plaquetas/efeitos dos fármacos , Plaquetas/efeitos da radiação , Carboplatina/toxicidade , Quimiorradioterapia/efeitos adversos , Modelos Biológicos , Modelos Estatísticos , Lesões por Radiação/induzido quimicamente , Trombocitopenia/induzido quimicamente , Irradiação Corporal Total/efeitos adversos , Animais , Antineoplásicos/administração & dosagem , Biomarcadores/sangue , Plaquetas/metabolismo , Plaquetas/patologia , Carboplatina/administração & dosagem , Simulação por Computador , Esquema de Medicação , Feminino , Humanos , Camundongos , Contagem de Plaquetas , Doses de Radiação , Lesões por Radiação/sangue , Receptores de Trombopoetina/sangue , Medição de Risco , Trombocitopenia/sangue , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação , Trombopoetina/sangue , Fatores de Tempo
7.
J Theor Biol ; 383: 44-60, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26232694

RESUMO

A mathematical model that describes the effects of acute radiation exposure on thrombopoiesis in primates and humans is presented. Thrombopoiesis is a complex multistage dynamic process with potential differences between species. Due to known differences in cellular radiosensitivities, nadir times, and cytopenia durations, direct extrapolation from rhesus to human platelet dynamics is unrealistic. Developing mathematical models of thrombopoiesis for both humans and primates allows for the comparison of the system's response across species. Thus, data obtained in primate experiments can be extrapolated to predictions in humans. Parameter values for rhesus macaques and humans were obtained either from direct experimental measurements or through optimization procedures using dynamic data on platelet counts following radiation exposure. Model simulations accurately predict trends observed in platelet dynamics: at low radiation doses platelet counts decline after a time lag, and nadir depth is dose dependent. The models were validated using data that was not used during the parameterization process. In particular, additional experimental data was used for rhesus, and accident and platelet donor data was used for humans. The model aims to simulate the average response in rhesus and humans following irradiation. Variation in platelet dynamics due to individual variability can be modeled using Monte Carlo simulations in which parameter values are sampled from distributions. This model provides insight into the time course of the physiological effects of radiation exposure, information which could be valuable for disaster planning and survivability analysis and help in drug development of radiation medical countermeasures.


Assuntos
Modelos Biológicos , Lesões por Radiação/fisiopatologia , Trombopoese/efeitos da radiação , Algoritmos , Animais , Relação Dose-Resposta à Radiação , Humanos , Macaca mulatta , Lesões por Radiação/patologia
8.
Platelets ; 26(5): 459-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25025394

RESUMO

Myelodysplastic syndromes (MDS) are hallmarked by cytopenia and dysplasia of hematopoietic cells, often accompanied by mitochondrial dysfunction and increases of reactive oxygen species (ROS) within affected cells. However, it is not known whether the increase in ROS production is an instigator or a byproduct of the disease. The present investigation shows that mice lacking immediate early responsive gene X-1 (IEX-1) exhibit lineage specific increases in ROS production and abnormal cytology upon radiation in blood cell types commonly identified in MDS. These affected cell lineages chiefly have the bone marrow as a primary site of differentiation and maturation, while cells with extramedullary differentiation and maturation like B- and T-cells remain unaffected. Increased ROS production is likely to contribute significantly to irradiation-induced thrombocytopenia in the absence of IEX-1 as demonstrated by effective reversal of the disorder after mitoquinone (MitoQ) treatment, a mitochondria-specific antioxidant. MitoQ reduced intracellular ROS production within megakaryocytes and platelets. It also normalized mitochondrial membrane potential and superoxide production in platelets in irradiated, IEX-1 deficient mice. The lineage-specific effects of mitochondrial ROS may help us understand the etiology of thrombocytopenia in association with MDS in a subgroup of the patients.


Assuntos
Compostos Organofosforados/farmacologia , Trombocitopenia/sangue , Trombocitopenia/etiologia , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação , Ubiquinona/análogos & derivados , Animais , Antioxidantes/metabolismo , Plaquetas/metabolismo , Medula Óssea/metabolismo , Linhagem da Célula/genética , Modelos Animais de Doenças , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Megacariócitos/efeitos da radiação , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Trombocitopenia/tratamento farmacológico , Ubiquinona/farmacologia , Irradiação Corporal Total
9.
J Nucl Med ; 55(8): 1355-60, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24935991

RESUMO

UNLABELLED: In targeted radionuclide radiotherapy, the relationship between bone marrow (BM) toxicity and absorbed dose seems to be elusive. A compartmental model of mouse thrombopoiesis and erythropoiesis was set up to predict the depletion of hematopoietic cells as a function of the irradiation dose delivered to BM by injected radiopharmaceuticals. All simulated kinetics were compared with experimental toxicity for several stages of differentiation of the 2 hematopoietic lineages. METHODS: C57BL/6 mice were injected either with (18)FNa (37 and 60 MBq), a bone-seeking agent, or with saline. BM mean absorbed doses were calculated according to the MIRD formalism from small-animal PET/CT images. Hematologic toxicity was monitored over time, after (18)FNa injection, by studying BM progenitors and precursors in addition to blood cells. The compartmental model takes into account the pharmacokinetics of the compound, in addition to cellular kinetics and cell radiosensitivities for the 2 studied lineages. RESULTS: Because biodistribution studies showed an uptake of (18)FNa in bones, the skeleton was considered as the principal source organ of BM irradiation. The time-activity curve obtained from validated quantification of PET/CT images allowed for the calculation of mean absorbed doses to the whole BM of 2.1 and 3.4 Gy for (18)FNa injections of 37 and 60 MBq, respectively. Concerning hematologic toxicity, the model was in good agreement for the 2 absorbed doses with experimental measurements of cell depletion for platelets, progenitors, and precursors within the BM in terms of time to nadir, depletion intensity, and time to recovery. The same agreement was obtained for red blood cells and their precursors. Model predictions demonstrated that BM toxicity was in correlation with the mean absorbed dose as higher depletions at nadir and longer delays to recovery were noticed for 3.4 Gy than for 2.1 Gy. CONCLUSION: The developed compartmental model of thrombopoiesis and erythropoiesis in a BM toxicity context, after internal irradiation, allowed for the prediction of cell kinetics of BM progenitors, precursors, and mature blood cells in a dose-dependent manner. This model could therefore be used to predict hematologic toxicity in preclinical internal radiotherapy to study the dose-response relationship.


Assuntos
Medula Óssea/efeitos da radiação , Eritropoese/efeitos da radiação , Modelos Biológicos , Trombopoese/efeitos da radiação , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Relação Dose-Resposta à Radiação , Feminino , Cinética , Camundongos , Radiometria
10.
Blood ; 124(2): 277-86, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24735964

RESUMO

Megakaryocyte (MK) development in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine stromal cell-derived factor-1 (SDF-1), signaling through CXCR4, is implicated in the maturational chemotaxis of MKs toward sinusoidal vessels. Here, we demonstrate that both IV administration of SDF-1 and stabilization of endogenous SDF-1 acutely increase MK-vasculature association and thrombopoiesis with no change in MK number. In the setting of radiation injury, we find dynamic fluctuations in marrow SDF-1 distribution that spatially and temporally correlate with variations in MK niche occupancy. Stabilization of altered SDF-1 gradients directly affects MK location. Importantly, these SDF-1-mediated changes have functional consequences for platelet production, as the movement of MKs away from the vasculature decreases circulating platelets, while MK association with the vasculature increases circulating platelets. Finally, we demonstrate that manipulation of SDF-1 gradients can improve radiation-induced thrombocytopenia in a manner additive with earlier TPO treatment. Taken together, our data support the concept that SDF-1 regulates the spatial distribution of MKs in the marrow and consequently circulating platelet numbers. This knowledge of the microenvironmental regulation of the MK lineage could lead to improved therapeutic strategies for thrombocytopenia.


Assuntos
Movimento Celular , Quimiocina CXCL12/fisiologia , Megacariócitos/citologia , Megacariócitos/fisiologia , Lesões Experimentais por Radiação , Nicho de Células-Tronco/genética , Trombopoese/genética , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Células da Medula Óssea/efeitos da radiação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Movimento Celular/efeitos da radiação , Células Cultivadas , Quimiocina CXCL12/administração & dosagem , Feminino , Células Progenitoras de Megacariócitos/citologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos/fisiologia , Células Progenitoras de Megacariócitos/efeitos da radiação , Megacariócitos/efeitos dos fármacos , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Lesões Experimentais por Radiação/genética , Lesões Experimentais por Radiação/patologia , Receptores CXCR4/administração & dosagem , Receptores CXCR4/metabolismo , Nicho de Células-Tronco/efeitos dos fármacos , Nicho de Células-Tronco/efeitos da radiação , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação
11.
Mol Med Rep ; 9(5): 1629-33, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24626603

RESUMO

The objective of the present study was to investigate the role of the steroid receptor coactivator-3 (SRC-3) in hematopoiesis of mouse bone marrow (BM) following total body irradiation (TBI). SRC-3-/­ mice and wild-type (WT) mice were exposed to 4.5 Gy γ rays. Immunoblotting analysis revealed that the SRC-3 protein (p160) levels in normal BM-nucleated cells in WT were higher than in SRC-3-/­ mice. Furthermore, peripheral blood cell counts, BM cellularity and colony-forming unit (CFU) assays were performed following irradiation. The results showed that peripheral blood cells were significantly lower in number and recovered less rapidly in irradiated SRC-3-/­ mice as compared with control animals. BM-nucleated cell and CFU counts were significantly decreased in SRC-3-/­ mice on the 7th and 14th day. Of note, the recovery of platelet (PLT) and megakaryocytic lineage were more depressed than the granulocytic and erythroid lineage in SRC-3-/­ mice. In conclusion, the present study demonstrated that the hematopoietic ability in SRC-3 knockout mice is severely impaired following a sublethal dose of irradiation.


Assuntos
Hematopoese/genética , Hematopoese/efeitos da radiação , Coativador 3 de Receptor Nuclear/genética , Trombopoese/genética , Trombopoese/efeitos da radiação , Irradiação Corporal Total , Animais , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/metabolismo , Células Sanguíneas/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Feminino , Megacariócitos/metabolismo , Megacariócitos/efeitos da radiação , Camundongos , Camundongos Knockout , Coativador 3 de Receptor Nuclear/deficiência
12.
PLoS One ; 8(3): e59385, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555027

RESUMO

Exposure of hematopoietic stem/progenitor cells (HSPCs) to ionizing radiation causes a marked suppression of mature functional blood cell production in a linear energy transfer (LET)- and/or dose-dependent manner. However, little information about LET effects on the proliferation and differentiation of HSPCs has been reported. With the aim of characterizing the effects of different types of LET radiations on human myeloid hematopoiesis, in vitro hematopoiesis in Human CD34(+) cells exposed to carbon-ion beams or X-rays was compared. Highly purified CD34(+) cells exposed to each form of radiation were plated onto semi-solid culture for a myeloid progenitor assay. The surviving fractions of total myeloid progenitors, colony-forming cells (CFC), exposed to carbon-ion beams were significantly lower than of those exposed to X-rays, indicating that CFCs are more sensitive to carbon-ion beams (D(0) = 0.65) than to X-rays (D(0) = 1.07). Similar sensitivities were observed in granulocyte-macrophage and erythroid progenitors, respectively. However, the sensitivities of mixed-type progenitors to both radiation types were similar. In liquid culture for 14 days, no significant difference in total numbers of mononuclear cells was observed between non-irradiated control culture and cells exposed to 0.5 Gy X-rays, whereas 0.5 Gy carbon-ion beams suppressed cell proliferation to 4.9% of the control, a level similar to that for cells exposed to 1.5 Gy X-rays. Cell surface antigens associated with terminal maturation, such as CD13, CD14, and CD15, on harvest from the culture of X-ray-exposed cells were almost the same as those from the non-irradiated control culture. X-rays increased the CD235a(+) erythroid-related fraction, whereas carbon-ion beams increased the CD34(+)CD38(-) primitive cell fraction and the CD13(+)CD14(+/-)CD15(-) fraction. These results suggest that carbon-ion beams inflict severe damage on the clonal growth of myeloid HSPCs, although the intensity of cell surface antigen expression by mature myeloid cells derived from HSPCs exposed to each type of radiation was similar to that by controls.


Assuntos
Diferenciação Celular/efeitos da radiação , Sangue Fetal/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos da radiação , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Proliferação de Células/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Sangue Fetal/citologia , Íons Pesados , Hematopoese/efeitos da radiação , Células-Tronco Hematopoéticas/citologia , Humanos , Transferência Linear de Energia , Trombopoese/efeitos da radiação , Raios X
13.
Exp Hematol ; 41(7): 635-645.e3, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23507524

RESUMO

Sublethal irradiation and 5-fluorouracil (5-FU) treatment are two commonly used myelosuppressive methods used in the study of hematopoiesis. These methods have been considered interchangeable by some researchers because the morphological changes in the bone marrow to these treatments are similar. Here, we sought to compare the responses of hematopoietic cells, stem and progenitor cells and the bone marrow microenvironment to these treatments. Although bone marrow cellularity decreased after both treatments, the underlying mechanism of the bone marrow cell regression and recovery were very different between the two models. We found: 1. Myeloid cells and lymphoid cells had different sensitivity to the different treatments. 2. Following an initial decrease in stem cell number, 5-FU treated mice had profound thrombopoietin (Tpo) dependent stem cell rebound above baseline levels. 3. Platelet rebound in 5-FU treated animals was not the result of stem cell rebound. 4. Stem cell and platelet rebound did not occur in sub-lethally irradiated mice. 5. Platelet rebound resulted from an indirect effect of 5-FU on the microenvironment cells, but not a direct effect on the stem cells. 6. Microarray studies demonstrated that up-regulation of the angiopoietin-1/Tie2 signaling pathway coincided with platelet rebound. 7. Suppression of genes involved in chromosomal organization coincided with stem cell and platelet rebound.


Assuntos
Plaquetas/efeitos dos fármacos , Fluoruracila/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Angiopoietina-1/biossíntese , Angiopoietina-1/genética , Animais , Plaquetas/efeitos da radiação , Medula Óssea/efeitos dos fármacos , Contagem de Células , Divisão Celular/efeitos dos fármacos , Microambiente Celular , Estruturas Cromossômicas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hematopoese/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Análise em Microsséries , Quimera por Radiação , Lesões Experimentais por Radiação/patologia , Lesões Experimentais por Radiação/fisiopatologia , Receptor TIE-2/biossíntese , Receptor TIE-2/genética , Receptores de Trombopoetina/deficiência , Receptores de Trombopoetina/genética , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação , Trombopoetina/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
Health Phys ; 103(6): 787-801, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23111526

RESUMO

Biologically motivated mathematical models, which describe the dynamics of the thrombocytopoietic, granulocytopoietic, and erythropoietic systems in irradiated humans, are thoroughly investigated. These models are the systems of nonlinear ordinary differential equations, whose variables and constant parameters have clear biological meaning. The modeling studies reveal general regularities and peculiarities of the dynamics of the aforementioned hematopoietic lines in acutely and chronically irradiated humans. It is shown that the predictions of the models qualitatively and quantitatively agree with the respective clinical data for humans exposed to acute and chronic irradiation in wide ranges of doses and dose rates. Moreover, the "lethal" dose rate of chronic irradiation, which is evaluated in the framework of the granulocytopoiesis model, coincides with the real minimal dose rate of lethal chronic irradiation for humans. As for the thrombocytopoiesis and erythropoiesis models, the respective "lethal" dose rates of chronic irradiation are very close to the real one for humans. All this bears witness to the validity of employment of the developed models in the investigation and prediction of radiation effects on human hematopoiesis.


Assuntos
Exposição Ambiental/efeitos adversos , Eritropoese/efeitos da radiação , Granulócitos/citologia , Granulócitos/efeitos da radiação , Modelos Biológicos , Trombopoese/efeitos da radiação , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Fatores de Tempo
15.
J Radiat Res ; 53(4): 581-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22843623

RESUMO

It is unclear how to effectively mitigate against irradiation injury. In this study, we studied the capacity of recombinant human insulin-like growth factor-I (rhIGF-I) on hematologic recovery in irradiated BALB/c mice and its possible mechanism. BALB/c mice were injected with rhIGF-I subcutaneously at a dose of 100 µg/kg twice daily for 7 days after total body irradiation. Compared with a saline control group, treatment with rhIGF-I significantly improved the survival of mice after lethal irradiation (7.5 Gy). It was found that treatment with rhIGF-I not only could increase the frequency of Sca-1(+) cells in bone marrow harvested at Day 14 after irradiation, but also it could decrease the apoptosis of mononuclear cells induced by irradiation as measured by flow cytometry, suggesting that rhIGF-I may mediate its effects primarily through promoting hematopoietic stem cell/progenitor survival and protecting mononuclear cells from apoptosis after irradiation exposure. Moreover, we have found that rhIGF-I might facilitate thrombopoiesis in an indirect way. Our data demonstrated that rhIGF-I could promote overall hematopoietic recovery after ionizing radiation and reduce the mortality when administered immediately post lethal irradiation exposure.


Assuntos
Células-Tronco Hematopoéticas/citologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Citometria de Fluxo/métodos , Injeções Subcutâneas , Leucócitos Mononucleares/patologia , Leucócitos Mononucleares/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tolerância a Radiação , Radiação Ionizante , Trombopoese/efeitos dos fármacos , Trombopoese/efeitos da radiação , Fatores de Tempo
16.
Radiat Res ; 176(6): 716-24, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22026586

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Trombopoese/efeitos da radiação , Antígenos CD34/metabolismo , Plaquetas/citologia , Plaquetas/efeitos da radiação , Feminino , Sangue Fetal/citologia , Regulação da Expressão Gênica/efeitos da radiação , Células-Tronco Hematopoéticas/metabolismo , Humanos , Interleucina-3/metabolismo , Megacariócitos/citologia , Megacariócitos/efeitos da radiação , Placenta/citologia , Placenta/efeitos da radiação , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Doses de Radiação , Trombopoetina/metabolismo
17.
Int J Radiat Oncol Biol Phys ; 80(5): 1533-40, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21740995

RESUMO

PURPOSE: Factors affecting the severity of radiation-induced thrombocytopenia (RIT) are not well described. We address whether platelet factor 4 (PF4; a negative paracrine for megakaryopoiesis) affects platelet recovery postradiation. METHODS AND MATERIALS: Using conditioned media from irradiated bone marrow (BM) cells from transgenic mice overexpressing human (h) PF4 (hPF4+), megakaryocyte colony formation was assessed in the presence of this conditioned media and PF4 blocking agents. In a model of radiation-induced thrombocytopenia, irradiated mice with varying PF4 expression levels were treated with anti-hPF4 and/or thrombopoietin (TPO), and platelet count recovery and survival were examined. RESULTS: Conditioned media from irradiated BM from hPF4+ mice inhibited megakaryocyte colony formation, suggesting that PF4 is a negative paracrine released in RIT. Blocking with an anti-hPF4 antibody restored colony formation of BM grown in the presence of hPF4+ irradiated media, as did antibodies that block the megakaryocyte receptor for PF4, low-density lipoprotein receptor-related protein 1 (LRP1). Irradiated PF4 knockout mice had higher nadir platelet counts than irradiated hPF4+/knockout litter mates (651 vs. 328 × 106/mcL, p = 0.02) and recovered earlier (15 days vs. 22 days, respectively, p <0.02). When irradiated hPF4+ mice were treated with anti-hPF4 antibody and/or TPO, they showed less severe thrombocytopenia than untreated mice, with improved survival and time to platelet recovery, but no additive effect was seen. CONCLUSIONS: Our studies show that in RIT, damaged megakaryocytes release PF4 locally, inhibiting platelet recovery. Blocking PF4 enhances recovery while released PF4 from megakaryocytes limits TPO efficacy, potentially because of increased release of PF4 stimulated by TPO. The clinical value of blocking this negative paracrine pathway post-RIT remains to be determined.


Assuntos
Plaquetas/efeitos da radiação , Megacariócitos/efeitos da radiação , Fator Plaquetário 4/fisiologia , Trombocitopenia/etiologia , Animais , Plaquetas/citologia , Medula Óssea/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias/métodos , Meios de Cultivo Condicionados/efeitos da radiação , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contagem de Plaquetas , Fator Plaquetário 4/antagonistas & inibidores , Receptores de LDL/antagonistas & inibidores , Trombocitopenia/sangue , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Trombopoese/fisiologia , Trombopoese/efeitos da radiação , Trombopoetina/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores
18.
Radiat Res ; 176(1): 8-16, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21529146

RESUMO

Hematopoietic processes, especially megakaryocytopoiesis and thrombopoiesis, are highly sensitive to high-linear energy transfer (LET) radiations such as heavy-ion beams that have greater biological effects than low-LET radiation. This study examined the terminal maturation of megakaryocytes and platelet production derived from hematopoietic stem cells irradiated with heavy-ion beams. CD34(+) cells derived from human placental/umbilical cord blood were exposed to monoenergetic carbon-ion beams (LET  =  50 keV/µm) and then cultured in a serum-free medium supplemented with thrombopoietin and interleukin-3. There was no significant difference in megakaryocyte-specific markers between nonirradiated control and irradiated cells. Expression of Tie-2, a receptor that acts in early hematopoiesis, showed a significant 1.31-fold increase after 2 Gy irradiation compared to control cells on day 7. There was a significant increase in Tie-2 mRNA expression. In addition, the expression of other mRNAs, such as PECAM1, SELP and CD44, was also significantly increased in cells irradiated with heavy-ion beams. However, the adherent function of platelets derived from the irradiated cells showed no difference from that in the controls. These results clarify that the functions of megakaryocytopoiesis and thrombopoiesis derived from hematopoietic stem/progenitor cells irradiated with heavy-ion beams are similar to those in the unirradiated cells, although heavy-ion beams affect the expression of genes associated with cellular adhesion.


Assuntos
Íons Pesados , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Megacariócitos/citologia , Megacariócitos/efeitos da radiação , Trombopoese/efeitos da radiação , Antígenos CD34/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Relação Dose-Resposta à Radiação , Sangue Fetal/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Células-Tronco Hematopoéticas/metabolismo , Humanos , Interleucina-3/farmacologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Testes de Função Plaquetária , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trombopoese/efeitos dos fármacos , Trombopoetina/farmacologia
19.
Transfusion ; 50(6): 1285-94, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20088841

RESUMO

BACKGROUND: We found when using a mouse model of immune thrombocytopenia (ITP) that platelet (PLT) nadir could not be maintained in the face of daily PLT antibody, making interpretation of treatment modalities difficult. This finding was documented to be at least in part due to increased thrombopoiesis as a result of a compensated thrombocytolytic state. Thus, it was important to develop an improved mouse model of human ITP so as to maintain PLT nadir over time. STUDY DESIGN AND METHODS: To maintain PLT nadir, we have developed two mouse models. One model uses single-dose sublethal total body gamma irradiation (TBI) in combination with daily low-dose PLT antibody administration while the second model uses escalation of the dose of PLT antibody over time. Both models maintain PLT nadir and allow for the study of treatment modalities without interference by marrow compensation. RESULTS: Surprisingly, intravenous immune globulin (IVIG) shows no efficacy when using the TBI combination model but works well using the dose-escalation mouse model. In contrast, anti-TER-119 shows efficacy using either mouse model. Our results indicate that the mechanism of action of IVIG requires a functional marrow and/or involves a radiosensitive regulatory cell. However, IVIG works using the dose-escalation model without TBI and the increase in PLT counts correlates directly with reticulated PLTs suggesting that the IVIG mechanism involves effects on megakaryopoiesis/thrombopoiesis. CONCLUSIONS: These mouse models should be useful for investigators wishing to maintain PLT nadir over prolonged periods of time for the study of mechanism and efficacy of various treatments for ITP.


Assuntos
Plaquetas/imunologia , Modelos Animais de Doenças , Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Animais , Feminino , Raios gama , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Púrpura Trombocitopênica Idiopática/induzido quimicamente , Púrpura Trombocitopênica Idiopática/imunologia , Trombopoese/imunologia , Trombopoese/efeitos da radiação , Fatores de Tempo , Irradiação Corporal Total
20.
J Radiat Res ; 50(5): 477-86, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19628925

RESUMO

Heavy ion beams are a high-LET radiation that has greater biological effect than electron beams or X-rays. However, little is known about the effect of heavy ion beams on the proliferation and differentiation of human hematopoietic stem/progenitor cells (HSPCs). The present study examined the effect of heavy ion beams on gene expression in human HSPCs, especially during early stage of megakaryocytopoiesis. Human CD34+ cells were exposed to monoenergetic carbon-ion beams (290 MeV/nucleon, LET = 50 KeV/m) that were generated by an accelerator (Heavy Ion Medical Accelerator in Chiba). The expression of various genes related to early hematopoiesis, megakaryocytopoiesis/erythropoiesis, cytokine receptors and oxidative stress were analyzed by real-time RT-PCR. Friend leukemia virus integration 1, an early hematopoiesis-related gene, showed significantly higher mRNA expression than the control at 6 hr after irradiation. In contrast, no significant differences were observed in almost all of the other early hematopoiesis-related genes, cytokine receptor-coded genes and megakaryocytopoiesis/erythropoiesis-differentiation pathway-related genes, respectively. An analysis of the response of the genes to oxidative stress revealed the expression of heme oxygenase 1 to show a 1.5-fold and 11.9-fold increase from the day 0 control at 24 hr after 0.5 Gy and 2 Gy irradiation, respectively. Similarly, the NAD(P)H dehydrogenase-quinone 1 expression also showed a 22.0-fold and a 21.8-fold increase at 6 hr in comparison to the initial control. These results showed that the heavy ion beams affect megakaryocytopoiesis/ erythropoiesis differentiation of human HSPCs on the gene expression level.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , RNA Mensageiro/metabolismo , Trombopoese/fisiologia , Trombopoese/efeitos da radiação , Células Cultivadas , Íons Pesados , Células-Tronco Hematopoéticas/citologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA