Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673041

RESUMO

Primary thrombopoietic mediator thrombopoietin (THPO) is mainly produced by the liver; it may act as a growth factor for hepatic progenitors. Principal angiogenesis inducer vascular endothelial growth factor-A (VEGF-A) is critical for the complex vascular network within the liver architecture. As a cross-regulatory loop between THPO and VEGF-A has been demonstrated in the hematopoietic system, the two growth factors were hypothesized to cooperatively contribute to the progression from liver cirrhosis (LC) to hepatocellular carcinoma (HCC). The mRNA and protein expression levels of THPO, VEGF-A, and their receptors were examined, compared, and correlated in paired cancerous and LC tissues from 26 cirrhosis-related HCC patients, using qRT-PCR and immunohistochemistry. THPO and VEGF-A were alternatively silenced by small interfering RNA (siRNA) in human liver cancer cell lines Huh7 and HepG2. THPO and VEGF-A expressions significantly increased in tumor versus LC tissues. HCC and paired LC cells expressed similar levels of THPO receptor (R), whereas vascular endothelial growth factor receptor (VEGFR) -1 and VEGFR-2 levels were higher in HCC than in corresponding LC tissue samples. A significant linear correlation emerged between THPO and VEGF-A transcripts in HCC and, at the protein level, THPO and THPOR were significantly correlated with VEGF-A in tumor tissues. Both HCC and LC expressed similar levels of gene and protein hypoxia inducible factor (HIF)-1α. Positive cross-regulation occurred with the alternative administration of siRNAs targeting THPO and those targeting VEGF-A in hypoxic liver cancer cell lines. These results suggest THPO and VEGF-A might act as interdependently regulated autocrine and/or paracrine systems for cellular growth in HCC. This might be clinically interesting, since new classes of THPOR agonistic/antagonistic drugs may provide novel therapeutic options to correct the frequent hemostatic abnormality seen in HCC patients.


Assuntos
Carcinoma Hepatocelular/metabolismo , Cirrose Hepática/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/biossíntese , Trombopoetina/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Idoso , Comunicação Autócrina , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Cirrose Hepática/complicações , Cirrose Hepática/patologia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Comunicação Parácrina
2.
Hepatology ; 74(1): 411-427, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33369745

RESUMO

BACKGROUND AND AIMS: Thrombocytopenia has been described in most patients with acute and chronic liver failure. Decreased platelet production and decreased half-life of platelets might be a consequence of low levels of thrombopoietin (TPO) in these patients. Platelet production is tightly regulated to avoid bleeding complications after vessel injury and can be enhanced under elevated platelet destruction as observed in liver disease. Thrombopoietin (TPO) is the primary regulator of platelet biogenesis and supports proliferation and differentiation of megakaryocytes. APPROACH AND RESULTS: Recent work provided evidence for the control of TPO mRNA expression in liver and bone marrow (BM) by scanning circulating platelets. The Ashwell-Morell receptor (AMR) was identified to bind desialylated platelets to regulate hepatic thrombopoietin (TPO) production by Janus kinase (JAK2)/signal transducer and activator of transcription (STAT3) activation. Two-thirds partial hepatectomy (PHx) was performed in mice. Platelet activation and clearance by AMR/JAK2/STAT3 signaling and TPO production were analyzed at different time points after PHx. Here, we demonstrate that PHx in mice led to thrombocytopenia and platelet activation defects leading to bleeding complications, but unaltered arterial thrombosis, in these mice. Platelet counts were rapidly restored by up-regulation and crosstalk of the AMR and the IL-6 receptor (IL-6R) to induce JAK2-STAT3-TPO activation in the liver, accompanied by an increased number of megakaryocytes in spleen and BM before liver was completely regenerated. CONCLUSIONS: The AMR/IL-6R-STAT3-TPO signaling pathway is an acute-phase response to liver injury to reconstitute hemostasis. Bleeding complications were attributable to thrombocytopenia and platelet defects induced by elevated PGI2 , NO, and bile acid plasma levels early after PHx that might also be causative for the high mortality in patients with liver disease.


Assuntos
Hepatectomia/efeitos adversos , Trombocitopenia/sangue , Trombopoetina/biossíntese , Animais , Receptor de Asialoglicoproteína/genética , Receptor de Asialoglicoproteína/metabolismo , Modelos Animais de Doenças , Humanos , Janus Quinase 2/metabolismo , Camundongos , Camundongos Knockout , Contagem de Plaquetas , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Organismos Livres de Patógenos Específicos , Trombocitopenia/etiologia , Trombopoetina/sangue
3.
Aging (Albany NY) ; 12(8): 7397-7410, 2020 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-32341206

RESUMO

Thrombopoietin (TPO) is a growth factor for the megakaryocytic/platelet lineage. In this study, we investigated the expression of TPO and its receptor, c-Mpl, in the human central nervous system (CNS) and their roles after a neural insult. Our results demonstrate that both TPO and c-Mpl are expressed in the neurons of the human CNS. TPO was also detected in human cerebrospinal fluid. TPO was found to be neuroprotective in hypoxic-ischemic neonatal rat brain models. In these rat models, treatment with TPO reduced brain damage and improved sensorimotor functions. In addition, TPO promoted C17.2 cell proliferation through activation of the PI3K/Akt signaling pathway. Via the Bcl-2/BAX signaling pathway, TPO exerted an antiapoptotic effect by suppressing mitochondrial membrane potentials. Taken together, our results indicate that TPO is neuroprotective in the CNS.


Assuntos
Sistema Nervoso Central/metabolismo , Regulação da Expressão Gênica , Neurônios/metabolismo , Receptores de Trombopoetina/genética , Trombopoetina/genética , Animais , Apoptose , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sistema Nervoso Central/citologia , Humanos , Modelos Animais , Neurônios/citologia , Ratos , Receptores de Trombopoetina/biossíntese , Transdução de Sinais , Trombopoetina/biossíntese
4.
Blood ; 135(25): 2292-2301, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32157300

RESUMO

Immune thrombocytopenia (ITP) is an acquired bleeding disorder characterized by antibody-mediated platelet destruction. Different mechanisms have been suggested to explain accelerated platelet clearance and impaired thrombopoiesis, but the pathophysiology of ITP has yet to be fully delineated. In this study, we tested 2 mouse models of immune-mediated thrombocytopenia using the rat anti-mouse GPIbα monoclonal antibody 5A7, generated in our laboratory. After a single IV administration of high-dose (2 mg/kg) 5A7, opsonized platelets were rapidly cleared from the circulation into the spleen and liver; this was associated with rapid upregulation of thrombopoietin (TPO) messenger RNA. In contrast, subcutaneous administration of low-dose 5A7 (0.08-0.16 mg/kg) every 3 days gradually lowered the platelet count; in this case, opsonized platelets were observed only in the spleen, and TPO levels remained unaltered. Interestingly, in both models, the 5A7 antibody was found on the surface of, as well as internalized to, bone marrow megakaryocytes. Consequently, platelets generated in the chronic phase of repeated subcutaneous 5A7 administration model showed reduced GPIbα membrane expression on their surface. Our findings indicate that evaluation of platelet surface GPIbα relative to platelet size may be a useful marker to support the diagnosis of anti-GPIbα antibody-induced ITP.


Assuntos
Anticorpos Monoclonais/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Púrpura Trombocitopênica Idiopática/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/toxicidade , Reações Antígeno-Anticorpo , Plaquetas/imunologia , Modelos Animais de Doenças , Injeções Intravenosas , Injeções Subcutâneas , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Opsonizantes/imunologia , Agregação Plaquetária/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Púrpura Trombocitopênica Idiopática/etiologia , RNA Mensageiro/biossíntese , Ratos , Baço/patologia , Trombopoetina/biossíntese , Trombopoetina/genética , Regulação para Cima
5.
J Transl Med ; 16(1): 199, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-30021591

RESUMO

BACKGROUND: Stored platelets undergo deleterious changes, referred to as platelet storage lesions (PSLs), which accelerate the desialylation of platelets and result in their phagocytosis and clearance by hepatic macrophages. Recent studies have reported that Ashwell-Morell receptor binds to desialylated platelets, thereby inducing hepatic thrombopoietin (TPO) production in a mouse model. Therefore, this study aimed to demonstrate these relationships between PSL and hepatic TPO production in human study. METHODS: Platelet concentrates (PCs) were obtained from 5 healthy volunteers and the remaining were discarded samples from the blood bank. PCs were divided into two halves, and stored either at 22 or 4 °C. Experiments were conducted using serial samples. Desialylation was assessed using flow cytometry, and structural changes were visualized using electron microscopy. Following co-culture of HepG2 cells (HB-8065, ATCC) with isolated platelets, hepatic TPO production was determined using real-time quantitative polymerase chain reaction and the supernatant TPO level was measured using a Luminex kit. RESULTS: For 5 days of storage duration, platelet counts were not influenced by the storage conditions, but the degree of desialylation was proportional to the storage duration. Significant changes in the platelet surface and structure according to storage conditions were noted in electron microscopy. HepG2 cells incubated with aged platelets expressed more TPO mRNA, and supernatant TPO levels were proportional to the storage duration. Refrigeration also influenced on the results of this study, but they were not statistically significant. CONCLUSIONS: This is the first study to demonstrate that, in vitro, aging and refrigeration affect the integrity of human platelets, resulting in induction of hepatic TPO mRNA and protein expression.


Assuntos
Plaquetas/metabolismo , Fígado/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Preservação Biológica , Temperatura , Trombopoetina/biossíntese , Acetilglucosamina/metabolismo , Adulto , Plaquetas/ultraestrutura , Citometria de Fluxo , Células Hep G2 , Humanos , Pessoa de Meia-Idade , Contagem de Plaquetas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trombopoetina/genética , Trombopoetina/metabolismo
6.
J Thromb Haemost ; 16(9): 1686-1699, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29975003

RESUMO

Platelets can contribute to tumor progression and metastasis. Cancer patients are at increased risk of thrombosis, and advanced stages of cancer are associated with thrombocytosis or increased platelet reactivity. Tyrosine kinase inhibitors (TKIs) are widely used as a targeted strategy for cancer treatment, with the aim of prolonging progression-free survival of the patients. Because of their broad kinase target spectrum, most TKIs inevitably have off-target effects. Platelets rely on tyrosine kinase activity for their activation. Frequently observed side effects are lowering of platelet count and inhibition of platelet functions, whether or not accompanied by an increased bleeding risk. In this review, we aim to give insights into: (i) 38 TKIs that are currently used for the treatment of different types of cancer, either on the market or in clinical trials; (ii) how distinct TKIs can inhibit activation mechanisms in platelets; and (iii) the clinical consequences of the antiplatelet effects of TKI treatment. For several TKIs, the knowledge on affinity for their targets does not align with the published effects on platelets and reported bleeding events. This review should raise awareness of the potential antiplatelet effects of several TKIs, which will be enhanced in the presence of antithrombotic drugs.


Assuntos
Antineoplásicos/farmacologia , Plaquetas/efeitos dos fármacos , Terapia de Alvo Molecular/efeitos adversos , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Trombofilia/etiologia , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos , Interleucina-6/biossíntese , Metástase Neoplásica , Proteínas de Neoplasias/fisiologia , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neovascularização Patológica/etiologia , Neovascularização Patológica/fisiopatologia , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/fisiologia , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/fisiologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/fisiologia , Trombocitose/etiologia , Trombofilia/induzido quimicamente , Trombofilia/prevenção & controle , Trombopoetina/biossíntese
7.
Blood ; 132(6): 622-634, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-29794068

RESUMO

Thrombopoietin (TPO), a hematopoietic growth factor produced predominantly by the liver, is essential for thrombopoiesis. Prevailing theory posits that circulating TPO levels are maintained through its clearance by platelets and megakaryocytes via surface c-Mpl receptor internalization. Interestingly, we found a two- to threefold decrease in circulating TPO in GPIbα-/- mice compared with wild-type (WT) controls, which was consistent in GPIbα-deficient human Bernard-Soulier syndrome (BSS) patients. We showed that lower TPO levels in GPIbα-deficient conditions were not due to increased TPO clearance by GPIbα-/- platelets but rather to decreased hepatic TPO mRNA transcription and production. We found that WT, but not GPIbα-/-, platelet transfusions rescued hepatic TPO mRNA and circulating TPO levels in GPIbα-/- mice. In vitro hepatocyte cocultures with platelets or GPIbα-coupled beads further confirm the disruption of platelet-mediated hepatic TPO generation in the absence of GPIbα. Treatment of GPIbα-/- platelets with neuraminidase caused significant desialylation; however, strikingly, desialylated GPIbα-/- platelets could not rescue impaired hepatic TPO production in vivo or in vitro, suggesting that GPIbα, independent of platelet desialylation, is a prerequisite for hepatic TPO generation. Additionally, impaired hepatic TPO production was recapitulated in interleukin-4/GPIbα-transgenic mice, as well as with antibodies targeting the extracellular portion of GPIbα, demonstrating that the N terminus of GPIbα is required for platelet-mediated hepatic TPO generation. These findings reveal a novel nonredundant regulatory role for platelets in hepatic TPO homeostasis, which improves our understanding of constitutive TPO regulation and has important implications in diseases related to GPIbα, such as BSS and auto- and alloimmune-mediated thrombocytopenias.


Assuntos
Síndrome de Bernard-Soulier/sangue , Plaquetas/fisiologia , Fígado/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Trombopoetina/biossíntese , Animais , Síndrome de Bernard-Soulier/genética , Células Cultivadas , Glicosilação , Hepatócitos/metabolismo , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico/metabolismo , Transfusão de Plaquetas , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/metabolismo , Trombopoetina/sangue
8.
Exp Hematol ; 51: 36-46, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28456746

RESUMO

Recently, NOD-SCID IL2Rγ-/- (NSG) mice were implanted with human mesenchymal stromal cells (MSCs) in the presence of ceramic scaffolds or Matrigel to mimic the human bone marrow (BM) microenvironment. This approach allowed the engraftment of leukemic samples that failed to engraft in NSG mice without humanized niches and resulted in a better preservation of leukemic stem cell self-renewal properties. To further improve our humanized niche scaffold model, we genetically engineered human MSCs to secrete human interleukin-3 (IL-3) and thrombopoietin (TPO). In vitro, these IL-3- and TPO-producing MSCs were superior in expanding human cord blood (CB) CD34+ hematopoietic stem/progenitor cells. MLL-AF9-transduced CB CD34+ cells could be transformed efficiently along myeloid or lymphoid lineages on IL-3- and TPO-producing MSCs. In vivo, these genetically engineered MSCs maintained their ability to differentiate into bone, adipocytes, and other stromal components. Upon transplantation of MLL-AF9-transduced CB CD34+ cells, acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) developed in engineered scaffolds, in which a significantly higher percentage of myeloid clones was observed in the mouse compartments compared with previous models. Engraftment of primary AML, B-cell ALL, and biphenotypic acute leukemia (BAL) patient samples was also evaluated, and all patient samples could engraft efficiently; the myeloid compartment of the BAL samples was better preserved in the human cytokine scaffold model. In conclusion, we show that we can genetically engineer the ectopic human BM microenvironment in a humanized scaffold xenograft model. This approach will be useful for functional study of the importance of niche factors in normal and malignant human hematopoiesis.


Assuntos
Diferenciação Celular , Engenharia Genética , Interleucina-3 , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco , Trombopoetina , Tecidos Suporte/química , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Interleucina-3/biossíntese , Interleucina-3/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Trombopoetina/biossíntese , Trombopoetina/genética
9.
Dev Comp Immunol ; 53(1): 13-22, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26111997

RESUMO

The use of in vitro colony assays in mammals has contributed to identification of erythroid progenitor cells such as burst-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E) progenitors, and serves to examine functions of erythropoietic growth factors like Erythropoietin (Epo) and Kit ligand. Here, we established an in vitro colony-forming assay capable of investigating erythropoiesis in carp (Cyprinus carpio), cloned and functionally characterized recombinant homologous molecules Epo and Kit ligand A (Kitla), and identified three distinct erythroid progenitor cells in carp. Recombinant carp Epo induced the formation of CFU-E-like and BFU-E-like erythroid colonies, expressing erythroid marker genes, ß-globin, epor and gata1. Recombinant carp Kitla alone induced limited colony formation, whereas a combination of Kitla and Epo dramatically enhanced erythroid colony formation and colony cell growth, as well as stimulated the formation of thrombocytic/erythroid colonies expressing not only erythroid markers but also thrombocytic markers, cd41 and c-mpl. Utilizing this colony assay to examine the distribution of distinct erythroid progenitor cells in carp, we demonstrated that carp head and trunk kidney play a primary role in erythropoiesis, while the spleen plays a secondary. Furthermore, we showed that presumably bi-potent thrombocytic/erythroid progenitor cells localize principally in the trunk kidney. Our results indicate that teleost fish possess mechanisms of Epo- and Kitla-dependent erythropoiesis similar to those in other vertebrates, and also help to demonstrate the diversity of erythropoietic sites among vertebrates.


Assuntos
Eritropoese/fisiologia , Eritropoetina/genética , Fator de Células-Tronco/genética , Células-Tronco/citologia , Animais , Carpas , Fator de Transcrição GATA1/biossíntese , Rim/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/biossíntese , Receptores da Eritropoetina/biossíntese , Proteínas Recombinantes/genética , Baço/metabolismo , Trombopoetina/biossíntese , Globinas beta/biossíntese
11.
Can J Gastroenterol Hepatol ; 28(10): 558-64, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25222481

RESUMO

Thrombocytopenia (defined as a platelet count <150×10(9)) is a well-known complication in patients with liver cirrhosis and has been observed in 76% to 85% of patients. Significant thrombocytopenia (platelet count <50×10(9) to 75×10(9)) occurs in approximately 13% of patients with cirrhosis. Thrombocytopenia can negatively impact the care of patients with severe liver disease by potentially interfering with diagnostic and therapeutic procedures. Multiple factors can contribute to the development of thrombocytopenia including splenic platelet sequestration, immunological processes, bone marrow suppression by chronic viral infection, and reduced levels or activity of the hematopoietic growth factor thrombopoietin. The present review focuses on the etiologies and management options for severe thrombocytopenia in the setting of advanced liver disease.


Assuntos
Cirrose Hepática/complicações , Hepatopatias/complicações , Trombocitopenia/terapia , Antivirais/uso terapêutico , Ablação por Cateter , Embolização Terapêutica , Feminino , Hepatite C/complicações , Humanos , Hiperesplenismo/complicações , Imunossupressores/uso terapêutico , Masculino , Derivação Portossistêmica Transjugular Intra-Hepática/métodos , Baço/fisiopatologia , Baço/cirurgia , Esplenectomia , Artéria Esplênica/cirurgia , Esplenomegalia/complicações , Trombocitopenia/complicações , Trombocitopenia/etiologia , Trombocitopenia/cirurgia , Trombopoetina/biossíntese
12.
J Acquir Immune Defic Syndr ; 65(5): 510-6, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24220290

RESUMO

BACKGROUND: Thrombocytopenia is a known consequence of HIV infection, and decreased production of platelets has been previously implicated in the pathogenesis of platelet decline during asymptomatic infection. Thrombopoietin (THPO) drives platelet production by stimulating the maturation of bone marrow megakaryocytes and can be transcriptionally downregulated by cytokines that are increased during infection such as transforming growth factor ß (TGFß) and platelet factor 4 (pf4). DESIGN: To determine whether transcriptional downregulation of THPO contributed to decreased platelet production during asymptomatic infection in the simian immunodeficiency virus (SIV)/macaque model of HIV, we compared hepatic THPO mRNA levels to platelet number and megakaryocyte density. To identify potential inhibitory factors that decrease THPO transcription during asymptomatic infection, we measured TGFß and pf4 plasma levels. To determine whether combined antiretroviral therapy (cART) could correct platelet decline by altering cytokine levels, we measured TGFß and pf4 in cART-treated SIV-infected macaques and compared these values to cART-untreated SIV-infected macaques. RESULTS: Hepatic THPO transcription was downregulated during asymptomatic SIV infection concurrent with platelet decline. Hepatic THPO mRNA levels correlated with bone marrow megakaryocyte density. In contrast, plasma TGFß levels were inversely correlated with hepatic THPO transcription and bone marrow megakaryocyte density. With cART treatment, plasma TGFß levels and platelet count returned to values similar to those in uninfected macaques. CONCLUSIONS: TGFß-mediated downregulation of hepatic THPO may lead to decline in platelet number during asymptomatic SIV infection, and cART may prevent platelet decline by normalizing plasma TGFß levels.


Assuntos
Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Trombocitopenia , Trombopoese , Trombopoetina/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Animais , Antirretrovirais/uso terapêutico , Doenças Assintomáticas , Regulação para Baixo , Perfilação da Expressão Gênica , Fígado/patologia , Macaca nemestrina , Masculino , Megacariócitos/fisiologia , Contagem de Plaquetas , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Trombopoetina/sangue , Resultado do Tratamento
13.
Proc Natl Acad Sci U S A ; 110(47): 18820-5, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24191050

RESUMO

Molecular targeting of the two receptor interaction domains of the epigenetic repressor silencing mediator of retinoid and thyroid hormone receptors (SMRT(mRID)) produced a transplantable skeletal syndrome that reduced radial bone growth, increased numbers of bone-resorbing periosteal osteoclasts, and increased bone fracture risk. Furthermore, SMRT(mRID) mice develop spontaneous primary myelofibrosis, a chronic, usually idiopathic disorder characterized by progressive bone marrow fibrosis. Frequently linked to polycythemia vera and chronic myeloid leukemia, myelofibrosis displays high patient morbidity and mortality, and current treatment is mostly palliative. To decipher the etiology of this disease, we identified the thrombopoietin (Tpo) gene as a target of the SMRT-retinoic acid receptor signaling pathway in bone marrow stromal cells. Chronic induction of Tpo in SMRT(mRID) mice results in up-regulation of TGF-ß and PDGF in megakaryocytes, uncontrolled proliferation of bone marrow reticular cells, and fibrosis of the marrow compartment. Of therapeutic relevance, we show that this syndrome can be rescued by retinoid antagonists, demonstrating that the physical interface between SMRT and retinoic acid receptor can be a potential therapeutic target to block primary myelofibrosis disease progression.


Assuntos
Medula Óssea/metabolismo , Citocinas/metabolismo , Repressão Epigenética/fisiologia , Correpressor 2 de Receptor Nuclear/antagonistas & inibidores , Mielofibrose Primária/tratamento farmacológico , Transdução de Sinais/fisiologia , Trombopoetina/genética , Fosfatase Alcalina/sangue , Animais , Benzotiazóis , Cálcio/sangue , Proliferação de Células/efeitos dos fármacos , Primers do DNA/genética , Diaminas , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Luciferases , Megacariócitos/metabolismo , Camundongos , Correpressor 2 de Receptor Nuclear/genética , Compostos Orgânicos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Reação em Cadeia da Polimerase , Mielofibrose Primária/etiologia , Quinolinas , Trombopoetina/biossíntese , Fator de Crescimento Transformador beta/metabolismo
14.
J Comp Pathol ; 149(2-3): 298-302, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23664426

RESUMO

Thrombopoietin (THPO) is the major cytokine that regulates megakaryopoiesis and platelet production. Several human and murine studies have demonstrated that THPO is primarily synthesized in the liver, but the kidney, spleen and bone marrow are also sites of expression. The aim of this study was to determine THPO mRNA levels in a range of canine tissues by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). Samples of bone marrow (n = 5), liver (n = 10), lung (n = 10), renal cortex (n = 10), renal medulla (n = 5) and spleen (n = 10) were obtained from 10 healthy, hound-cross dogs aged 6-8 months. The highest THPO mRNA levels were found in the liver, followed by the bone marrow, spleen, lung and kidney. There was a 13-fold difference in expression between liver and kidney. The bone marrow showed high levels of THPO mRNA in the absence of disease. The liver and bone marrow are likely to be the major sites of THPO production in the dog.


Assuntos
RNA Mensageiro/análise , Trombopoetina/biossíntese , Trombopoetina/genética , Transcriptoma , Animais , Medula Óssea/metabolismo , Cães , Fígado/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mediators Inflamm ; 2012: 390892, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22577249

RESUMO

Thrombopoietin (TPO) is a humoral growth factor originally identified for its ability to stimulate the proliferation and differentiation of megakaryocytes. In addition to its actions on thrombopoiesis, TPO directly modulates the homeostatic potential of mature platelets by influencing their response to several stimuli. In particular, TPO does not induce platelet aggregation per se but is able to enhance platelet aggregation in response to different agonists ("priming effect"). Our research group was actively involved, in the last years, in characterizing the effects of TPO in several human critical diseases. In particular, we found that TPO enhances platelet activation and monocyte-platelet interaction in patients with unstable angina, chronic cigarette smokers, and patients with burn injury and burn injury complicated with sepsis. Moreover, we showed that TPO negatively modulates myocardial contractility by stimulating its receptor c-Mpl on cardiomyocytes and the subsequent production of NO, and it mediates the cardiodepressant activity exerted in vitro by serum of septic shock patients by cooperating with TNF-α and IL-1ß. This paper will summarize the most recent results obtained by our research group on the pathogenic role of elevated TPO levels in these diseases and discuss them together with other recently published important studies on this topic.


Assuntos
Biomarcadores/metabolismo , Doenças Cardiovasculares/metabolismo , Trombopoetina/biossíntese , Angina Instável/metabolismo , Queimaduras/metabolismo , Proliferação de Células , Insuficiência Cardíaca/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-1beta/metabolismo , Óxido Nítrico/metabolismo , Ativação Plaquetária , Receptores de Trombopoetina/metabolismo , Traumatismo por Reperfusão/metabolismo , Choque Séptico/metabolismo , Fumar/efeitos adversos , Fator de Necrose Tumoral alfa/metabolismo
16.
Exp Hematol ; 39(3): 293-304, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21146580

RESUMO

OBJECTIVE: Our laboratory reported that the angiotensin converting enzyme inhibitor captopril improves erythroid recovery from total body irradiation (TBI) in mice when administered after irradiation. However, captopril administered before TBI attenuates erythroid recovery. Here we investigate captopril and radiation regulation of erythropoietin (EPO) and thrombopoietin (TPO), key effectors of erythroid progenitor proliferation and differentiation. MATERIALS AND METHODS: C57BL/6 mice, nonirradiated or exposed to 7.5 Gy TBI ((60)Co, 0.6 Gy/min) were untreated or administered captopril. Plasma EPO and TPO levels were measured by enzyme-linked immunosorbent assay. Gene expression of EPO was determined by quantitative reverse transcription polymerase chain reaction. The hypoxia-inducible factors (HIF)-1α and -2α were measured by immunoblotting. RESULTS: In nonirradiated mice, continuous captopril administration in the water transiently reduced reticulocytes and red blood cells after 7 and 10 days, respectively. EPO plasma levels and gene expression were reduced below detectable limits after 2 days of captopril treatment, but recovered within 7 days. HIF-1α and HIF-2α were activated preceding reticulocyte and red blood cell recovery. TBI, which ablates early and late-stage erythroid progenitors, activated both HIFs and increased EPO and TPO. Captopril treatment postirradiation suppressed radiation-induced HIF activation and EPO expression. In contrast, captopril administration for 7 days before TBI resulted in earlier EPO induction and activation. Captopril treatment lowered TPO levels in nonirradiated mice, but had minimal effects on radiation-induced TPO. CONCLUSIONS: In nonirradiated mice, captopril biphasically regulates EPO via HIF activation. TBI ablates erythroid progenitors, resulting in hypoxia, HIF activation, and increased EPO expression that are modulated by captopril treatment. These data suggest that short-term suppression of radiation-induced EPO immediately after TBI is favorable for erythroid recovery.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Captopril/farmacologia , Células Precursoras Eritroides/metabolismo , Eritropoetina/biossíntese , Raios gama/efeitos adversos , Regulação da Expressão Gênica , Irradiação Corporal Total , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Camundongos , Trombopoetina/biossíntese , Fatores de Tempo
17.
Proc Natl Acad Sci U S A ; 107(38): 16625-30, 2010 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20823251

RESUMO

With the notable exception of humans, uric acid is degraded to (S)-allantoin in a biochemical pathway catalyzed by urate oxidase, 5-hydroxyisourate (HIU) hydrolase, and 2-oxo-4-hydroxy-4-carboxy-5-ureidoimidazoline decarboxylase in most vertebrate species. A point mutation in the gene encoding mouse HIU hydrolase, Urah, that perturbed uric acid metabolism within the liver was discovered during a mutagenesis screen in mice. The predicted substitution of cysteine for tyrosine in a conserved helical region of the mutant-encoded HIU hydrolase resulted in undetectable protein expression. Mice homozygous for this mutation developed elevated platelet counts secondary to excess thrombopoietin production and hepatomegaly. The majority of homozygous mutant mice also developed hepatocellular carcinoma, and tumor development was accelerated by exposure to radiation. The development of hepatomegaly and liver tumors in mice lacking Urah suggests that uric acid metabolites may be toxic and that urate oxidase activity without HIU hydrolase function may affect liver growth and transformation. The absence of HIU hydrolase in humans predicts slowed metabolism of HIU after clinical administration of exogenous urate oxidase in conditions of uric acid-related pathology. The data suggest that prolonged urate oxidase therapy should be combined with careful assessment of toxicity associated with extrahepatic production of uric acid metabolites.


Assuntos
Amidoidrolases/deficiência , Amidoidrolases/genética , Hepatomegalia/enzimologia , Hepatomegalia/genética , Neoplasias Hepáticas Experimentais/enzimologia , Neoplasias Hepáticas Experimentais/genética , Mutação Puntual , Amidoidrolases/química , Amidoidrolases/metabolismo , Sequência de Aminoácidos , Animais , Feminino , Genes Supressores de Tumor , Hepatócitos/enzimologia , Hepatomegalia/etiologia , Neoplasias Hepáticas Experimentais/etiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Trombocitose/enzimologia , Trombocitose/genética , Trombopoetina/biossíntese , Urato Oxidase/metabolismo , Ácido Úrico/metabolismo , Ácido Úrico/toxicidade
18.
J Interferon Cytokine Res ; 30(7): 465-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20236022

RESUMO

Thrombopoietin (Tpo) and its receptor (c-Mpl; TpoR), which primary regulate megakaryopoiesis and platelet production, are also expressed in the central nervous system (CNS). Increased Tpo concentrations are present in the cerebrospinal fluid (CSF) of some patients with bacterial or viral meningitis. Since previous data implicated a proapoptotic role of Tpo on newly generated neuronal cells, we herein elucidated the regulation of Tpo in primary rat neurons (e17), astrocytes, and microglia (p0-p3), as well as in brain-derived vascular endothelial cells of 3-week-old rats after exposure to bacterial lipopolysaccharide (LPS). LPS inhibited Tpo gene expression in astrocytes and microglia, but not in neurons, most likely due to absence of Toll-like receptor 4 in neurons. While Tpo mRNA expression recovered in astrocytes after 24 h, it remained suppressed in microglia. Furthermore, we detected Tpo mRNA expression in primary brain-derived vascular endothelial cells, which also express the TpoR. In these cells, LPS significantly up-regulated Tpo mRNA expression. TpoR mRNA and protein expression remained constitutive in all cell types. Thus, our data provide evidence for a cell-type-specific modulation of Tpo mRNA expression by inflammation in brain-derived cells. Transient down-regulation of Tpo expression in astrocytes and microglia may limit Tpo-induced neuronal cell death in inflammatory brain disorders.


Assuntos
Astrócitos/metabolismo , Endotélio Vascular/metabolismo , Microglia/metabolismo , Receptores de Trombopoetina/biossíntese , Trombopoetina/biossíntese , Animais , Animais Recém-Nascidos , Astrócitos/imunologia , Astrócitos/patologia , Células Cultivadas , Regulação para Baixo , Embrião de Mamíferos , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Inflamação , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Microglia/imunologia , Microglia/patologia , Ratos , Ratos Wistar , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/imunologia , Trombopoetina/genética , Trombopoetina/imunologia , Regulação para Cima
19.
Artigo em Inglês | MEDLINE | ID: mdl-20008195

RESUMO

Major progress in understanding the pathogenesis in patients with thrombocytosis has been made by identifying mutations in the key regulators of thrombopoietin: the thrombopoietin receptor MPL and JAK2. Together, these mutations can be found in 50% to 60% of patients with essential thrombocythemia or primary myelofibrosis and in 10% to 20% of hereditary thrombocytosis. A decrease in expression of the Mpl protein can cause thrombocytosis even in the absence of mutations in the coding sequence, due to a shift in the balance between stimulation of signaling in megakaryopoiesis and removal of thrombopoietin by receptor mediated internalization in platelets. When present in a heterozygous state the JAK2-V617F mutation preferentially stimulates megakaryopoiesis and in most cases manifests as essential thrombocythemia (ET), whereas homozygous JAK2-V617F reduces megakaryopoiesis in favor of increased erythropoiesis, resulting in polycythemia vera and/or myelofibrosis. In 30% to 40% of patients with ET or primary myelofibrosis (PMF) and in 80% to 90% of pedigrees with hereditary thrombocytosis the disease-causing gene remains unknown. Ongoing genetic and genomic screens have identified genes that, when mutated, can cause thrombocytosis in mouse models. A more complete picture of the pathways that regulate megakaryopoisis and platelet production will be important for finding new ways of controlling platelet production in patients with thrombocytosis.


Assuntos
Janus Quinase 2/genética , Mutação , Receptores de Trombopoetina/genética , Trombocitose/genética , Trombopoetina/genética , Adulto , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Humanos , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Policitemia Vera/genética , Mielofibrose Primária/genética , Receptores de Trombopoetina/deficiência , Receptores de Trombopoetina/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Fatores de Transcrição STAT/fisiologia , Trombocitemia Essencial/genética , Trombopoese/genética , Trombopoetina/biossíntese , Trombopoetina/fisiologia , Transdução Genética
20.
Gynecol Obstet Invest ; 67(3): 145-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19023215

RESUMO

BACKGROUND: Following implantation, endometrial stroma is transformed into decidual tissue via a complex remodeling process. In parallel with that process, a significant increase in immune cells can be detected. Several studies suggest that chemokines and cytokines orchestrate the transformation of decidual tissue and the infiltration of leukocytes. In this study, we therefore compared chemokine and cytokine expression in the first- and third-trimester nonpregnant endometrium and decidua. METHODS: Investigation of the expression patterns of cytokines, chemokines and growth factors in endometrial tissue (after routine hysterectomy) and human decidual tissue (7-8 weeks of gestation and 38-40 weeks of gestation, respectively) was performed by protein array analysis. RESULTS: This analysis revealed a significant increase in monocyte-attracting chemokines (granulocyte-macrophage colony-stimulating factor, growth-related oncogene, growth-related oncogene-alpha and monocyte chemoattractant protein-2) , granulocyte-attracting chemokines (epithelial neutrophil activating peptide and interleukin 8) and proinflammatory factors (interleukin-1alpha, leptin) in decidual compared to endometrial tissue. Furthermore, concentrations of angiogenic substances (vascular endothelial growth factor and thrombopoietin) significantly peaked in first-trimester deciduas. CONCLUSION: This study demonstrates increased expression of chemokines and cytokines in decidual tissue compared to nonpregnant endometrium.


Assuntos
Citocinas/biossíntese , Endométrio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Regulação para Cima , Adulto , Quimiocinas/biossíntese , Decídua/metabolismo , Feminino , Idade Gestacional , Humanos , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Trombopoetina/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...