Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Hamostaseologie ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38925157

RESUMO

This review summarizes the rationale and current data on the use of thrombopoietin receptor agonists (TPO-RAs) for treating severe thrombocytopenia in infants, children, and adolescents. It focuses on substances that have been approved by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) for pediatric patients. Romiplostim and eltrombopag are already established as second-line treatment for persistent or chronic immune thrombocytopenia (ITP). As in adults, TPO-RAs are currently also evaluated in severe aplastic anemia (SAA), chemotherapy-induced thrombocytopenia (CIT), myelodysplastic syndromes (MDS), and poor engraftment after hematopoietic stem cell transplantation in pediatric and adolescent patients. Moreover, studies on the implication of TPO-RA in treating rare inherited thrombocytopenias, such as Wiskott-Aldrich syndrome (WAS), congenital amegakaryocytic thrombocytopenia (CAMT), or MYH9-associated thrombocytopenia, deserve future attention. Current developments include testing of avatrombopag and lusutrombopag that are approved for the treatment of thrombocytopenia associated with chronic liver disease (CLD) in adult patients. In pediatric and adolescent medicine, we expect in the near future a broader use of TPO-RAs as first-line treatment in primary ITP, thereby considering immunomodulatory effects that increase the rate of sustained remission off-treatment, and a selective use in rare inherited thrombocytopenias based on current clinical trials.

2.
Clin Genet ; 103(5): 585-589, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36562171

RESUMO

Known hereditary human diseases featuring impaired copper trafficking across cellular membranes involve ATP7A (Menkes disease, occipital horn disease, X-linked spinal muscular atrophy type 3) and ATP7B (Wilson disease). Herein, we report a newborn infant of consanguineous parents with a homozygous pathogenic variant in a highly conserved sequence of SLC31A1, coding for the copper influx transporter 1, CTR1. This missense variant, c.236T > C, was detected by whole exome sequencing. The infant was born with pulmonary hypoplasia and suffered from severe respiratory distress immediately after birth, necessitating aggressive mechanical ventilation. At 2 weeks of age, multifocal brain hemorrhages were diagnosed by cerebral ultrasound and magnetic resonance imaging, together with increased tortuosity of cerebral arteries. Ensuing seizures were only partly controlled by antiepileptic drugs, and the infant became progressively comatose. Laboratory investigations revealed very low serum concentrations of copper and ceruloplasmin. No hair shaft abnormalities were detected by dermatoscopy or light microscopic analyses of embedded hair shafts obtained at 4 weeks of life. The infant died after redirection of care and elective cessation of invasive mechanical ventilation at 1 month of age. This case adds SLC31A1 to the genes implicated in severe hereditary disorders of copper transport in humans.


Assuntos
Transportador de Cobre 1 , Degeneração Hepatolenticular , Síndrome dos Cabelos Torcidos , Humanos , Lactente , Recém-Nascido , Ceruloplasmina/genética , Cobre , Transportador de Cobre 1/genética , ATPases Transportadoras de Cobre/genética , Degeneração Hepatolenticular/genética , Síndrome dos Cabelos Torcidos/genética , Mutação de Sentido Incorreto
3.
Oxid Med Cell Longev ; 2022: 9714669, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35242281

RESUMO

During gestation, the most drastic change in oxygen supply occurs with the onset of ventilation after birth. As the too early exposure of premature infants to high arterial oxygen pressure leads to characteristic diseases, we studied the adaptation of the oxygen sensing system and its targets, the hypoxia-inducible factor- (HIF-) regulated genes (HRGs) in the developing lung. We draw a detailed picture of the oxygen sensing system by integrating information from qPCR, immunoblotting, in situ hybridization, and single-cell RNA sequencing data in ex vivo and in vivo models. HIF1α protein was completely destabilized with the onset of pulmonary ventilation, but did not coincide with expression changes in bona fide HRGs. We observed a modified composition of the HIF-PHD system from intrauterine to neonatal phases: Phd3 was significantly decreased, while Hif2a showed a strong increase and the Hif3a isoform Ipas exclusively peaked at P0. Colocalization studies point to the Hif1a-Phd1 axis as the main regulator of the HIF-PHD system in mouse lung development, complemented by the Hif3a-Phd3 axis during gestation. Hif3a isoform expression showed a stepwise adaptation during the periods of saccular and alveolar differentiation. With a strong hypoxic stimulus, lung ex vivo organ cultures displayed a functioning HIF system at every developmental stage. Approaches with systemic hypoxia or roxadustat treatment revealed only a limited in vivo response of HRGs. Understanding the interplay of the oxygen sensing system components during the transition from saccular to alveolar phases of lung development might help to counteract prematurity-associated diseases like bronchopulmonary dysplasia.


Assuntos
Adaptação Fisiológica/genética , Desenvolvimento Embrionário/genética , Hipóxia/genética , Hipóxia/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Organogênese/genética , Oxigênio/metabolismo , Transdução de Sinais/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Gravidez , RNA-Seq/métodos , Ratos Wistar , Análise de Célula Única/métodos
4.
Front Pediatr ; 9: 728607, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869097

RESUMO

Background and Objective: Intrauterine growth restriction (IUGR) poses additional challenges in extremely low gestational age newborns (ELGANs). We assessed disturbed hematopoiesis and morbidities associated with this disorder. Methods: This single-center retrospective case-control study compared perinatal hematological profiles, major morbidities, and mortality of 49 infants (gestational age <28 weeks, birth weight ≤ 3rd percentile, and compromised placental function) and 98 infants (birth weight >10th percentile) matched for gestational age, year, and sex. Results: IUGR-ELGANs had significantly elevated nucleated red blood cells and lower neutrophil and platelet counts at birth and on the third day of life. During the first week of life, IUGR-ELGANs received more red blood cell, platelet, and plasma transfusions and were more intensively treated with antibiotics. Rates of infections acquired during the first week (59.2 vs. 17.3%, p < 0.001), severe bronchopulmonary dysplasia or death (42.9 vs. 17.3%, p < 0.01), and mortality (36.7 vs. 7.1%, p < 0.001) were markedly elevated in IUGR-ELGANs, but not of hemorrhages or other morbidities. Conclusions: IUGR-ELGANs have high rates of acquired infections during the first week of life and display severe pulmonary morbidity leading to bronchopulmonary dysplasia or death. The high rate of transfusions observed in these infants warrants further scrutiny.

5.
J Pediatr Hematol Oncol ; 43(2): e255-e259, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32459720

RESUMO

This study aimed to examine whether the transfusion of donor blood products, abnormal coagulation or inflammation increase the risk of venous thromboembolism (VTE) associated with central venous catheters (CVC) in neonates. A retrospective case-control study including 25 neonates with CVC-associated VTE and tightly matched controls with CVC, but without VTE was performed. The frequency of (i) abnormal coagulation screens, (ii) increased inflammatory marker proteins before catheter insertion, or (iii) catheter-associated blood stream infection did not differ between cases and controls. No difference was found in the number or type of transfusions within the last day before VTE. However, the total number of transfusions in the time period between catheter placement and VTE diagnosis (median 6.5 d) was significantly higher (P<0.001) in cases (44 red blood cell, 61 plasma, and 18 platelet transfusions) compared with an equal median time period of 7 days postcatheter insertion in controls (26/24/11). In conclusion, intensive transfusion treatment (through a peripheral line) after CVC insertion was associated with a higher risk of VTE (odds ratio 7.58; 95% confidence interval, 0.84-68.46), suggesting that transfusion of adult donor blood products into the cellular and plasmatic hemostatic system of the neonate increases the risk for CVC-associated VTE.


Assuntos
Transfusão de Sangue/estatística & dados numéricos , Cateteres Venosos Centrais/efeitos adversos , Doadores de Tecidos/provisão & distribuição , Tromboembolia Venosa/etiologia , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Recém-Nascido , Masculino , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Tromboembolia Venosa/patologia
6.
Semin Fetal Neonatal Med ; 21(1): 10-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26712568

RESUMO

Neonatal thrombocytopenia is widespread in preterm and term neonates admitted to neonatal intensive care units, with up to one-third of infants demonstrating platelet counts <150 × 10(9)/L. Thrombocytopenia may arise from maternal, placental or fetal/neonatal origins featuring decreased platelet production, increased consumption, or both mechanisms. Over the past years, innovations in managing neonatal thrombocytopenia were achieved from prospectively obtained clinical data on thrombocytopenia and bleeding events, animal studies on platelet life span and production rate and clinical use of fully automated measurement of reticulated platelets (immature platelet fraction). This review summarizes the pathophysiology of neonatal thrombocytopenia, current management including platelet transfusion thresholds and recent developments in megakaryopoietic agents. Furthermore, we propose a novel index score for bleeding risk in thrombocytopenic neonates to facilitate clinician's decision-making when to transfuse platelets.


Assuntos
Transfusão de Plaquetas , Trombocitopenia/terapia , Benzoatos/uso terapêutico , Tomada de Decisão Clínica , Fármacos Hematológicos/uso terapêutico , Humanos , Hidrazinas/uso terapêutico , Recém-Nascido , Terapia Intensiva Neonatal , Transfusão de Plaquetas/métodos , Transfusão de Plaquetas/normas , Pirazóis/uso terapêutico , Receptores Fc/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Trombocitopenia/diagnóstico , Trombocitopenia/etiologia , Trombocitopenia/fisiopatologia , Trombopoese/fisiologia , Trombopoetina/uso terapêutico
7.
J Pediatr ; 167(1): 52-7.e1-3, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25863661

RESUMO

OBJECTIVE: To investigate the safety and short term outcome of high dose recombinant human erythropoietin (rhEpo) given shortly after birth and subsequently over the first 2 days for neuroprotection to very preterm infants. STUDY DESIGN: Randomized, double masked phase II trial. Preterm infants (gestational age 26 0/7-31 6/7 weeks) were given rhEpo (nt = 229; 3000 U/kg body weight) or NaCl 0.9% (nc = 214) intravenously at 3, 12-18, and 36-42 hours after birth. RESULTS: There were no relevant differences between the groups for short-term outcomes such as mortality, retinopathy of prematurity, intraventricular hemorrhage, sepsis, necrotizing enterocolitis, and bronchopulmonary dysplasia. At day 7-10, we found significantly higher hematocrit values, reticulocyte, and white blood cell counts, and a lower platelet count in the rhEpo group. CONCLUSIONS: Early high-dose rhEpo administration to very premature infants is safe and causes no excess in mortality or major adverse events. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00413946.


Assuntos
Deficiências do Desenvolvimento/prevenção & controle , Eritropoetina/administração & dosagem , Recém-Nascido Prematuro , Fármacos Neuroprotetores/administração & dosagem , Displasia Broncopulmonar/epidemiologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Enterocolite Necrosante/epidemiologia , Europa (Continente)/epidemiologia , Hematócrito , Humanos , Hipóxia-Isquemia Encefálica/epidemiologia , Recém-Nascido , Hemorragias Intracranianas/epidemiologia , Contagem de Leucócitos , Leucomalácia Periventricular/epidemiologia , Contagem de Plaquetas , Proteínas Recombinantes/administração & dosagem , Contagem de Reticulócitos , Retinopatia da Prematuridade/epidemiologia , Sepse/epidemiologia
8.
BMC Res Notes ; 7: 702, 2014 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25297646

RESUMO

BACKGROUND: Studies on liver regeneration following partial hepatectomy (PH) have identified several microRNAs (miRNAs) that show a regulated expression pattern. These studies involve major surgery to access the liver, which is known to have intrinsic effects on hepatic gene expression and may also affect miRNA screening results. We performed two-third PH or sham laparotomy (SL) in Wistar rats to investigate the effect of both procedures on miRNA expression in liver tissue and corresponding plasma samples by microarray and qRT-PCR analyses. As control groups, non-treated rats and rats undergoing anesthesia only were used. RESULTS: We found that 49 out of 323 miRNAs (15%) were significantly deregulated after PH in liver tissue 12 to 48 hours postoperatively (>20% change), while 45 miRNAs (14%) were deregulated following SL. Out of these miRNAs, 10 miRNAs were similarly deregulated after PH and SL, while one miRNA showed opposite regulation. In plasma, miRNA upregulation was observed for miR-133a and miR-133b following PH and SL, whereas miR-100 and miR-466c were similarly downregulated following anesthesia and surgery. CONCLUSIONS: We show that miRNAs are indeed regulated by sham laparotomy and anesthesia in rats. These findings illustrate the critical need for finding appropriate control groups in experimental surgery.


Assuntos
Anestesia , Hepatectomia , Fígado/metabolismo , MicroRNAs/sangue , Animais , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
10.
J Immunol ; 189(11): 5105-11, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23169863

RESUMO

Long-lived plasma cells survive in a protected microenvironment for years or even a lifetime and provide humoral memory by establishing persistent Ab titers. Long-lived autoreactive, malignant, and allergen-specific plasma cells are likewise protected in their survival niche and are refractory to immunosuppression, B cell depletion, and irradiation. Their elimination remains an essential therapeutic challenge. Recent data indicate that long-lived plasma cells reside in a multicomponent plasma cell niche with a stable mesenchymal and a dynamic hematopoietic component, both providing essential soluble and membrane-bound survival factors. Alternative niches with different hematopoietic cell components compensate fluctuations of single cell types but may also harbor distinct plasma cell subsets. In this Brief Review, we discuss conventional therapies in autoimmunity and multiple myeloma in comparison with novel drugs that target plasma cells and their niches. In the future, such strategies may enable the specific depletion of pathogenic plasma cells while leaving the protective humoral memory intact.


Assuntos
Autoimunidade/efeitos dos fármacos , Hipersensibilidade/patologia , Mieloma Múltiplo/patologia , Plasmócitos/patologia , Anticorpos Monoclonais/farmacologia , Autoanticorpos/biossíntese , Autoanticorpos/imunologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Humanos , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/imunologia , Tolerância Imunológica/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/imunologia , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Receptores de Citocinas/antagonistas & inibidores , Receptores de Citocinas/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
11.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1363-72, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21430077

RESUMO

The liver has the unique capacity to regenerate after surgical resection. However, the regulation of liver regeneration is not completely understood. Recent reports indicate an essential role for small noncoding microRNAs (miRNAs) in the regulation of hepatic development, carcinogenesis, and early regeneration. We hypothesized that miRNAs are critically involved in all phases of liver regeneration after partial hepatectomy. We performed miRNA microarray analyses after 70% partial hepatectomy in rats under isoflurane anesthesia at different time points (0 h to 5 days) and after sham laparotomy. Putative targets of differentially expressed miRNAs were determined using a bioinformatic approach. Two-dimensional (2D)-PAGE proteomic analyses and protein identification were performed on specimens at 0 and 24 h after resection. The temporal dynamics of liver regeneration were characterized by 5-bromo- 2-deoxyuridine, proliferating cell nuclear antigen, IL-6, and hepatocyte growth factor. We demonstrate that miRNA expression patterns changed during liver regeneration and that these changes were most evident during the peak of DNA replication at 24 h after resection. Expression of 13 miRNAs was significantly reduced 12-48 h after resection (>25% change), out of which downreguation was confirmed in isolated hepatocytes for 6 miRNAs at 24 h, whereas three miRNAs were significantly upregulated. Proteomic analysis revealed 65 upregulated proteins; among them, 23 represent putative targets of the differentially expressed miRNAs. We provide a temporal miRNA expression and proteomic dataset of the regenerating rat liver, which indicates a primary function for miRNA during the peak of DNA replication. These data will assist further functional studies on the role of miRNAs during liver regeneration.


Assuntos
Replicação do DNA/fisiologia , Hepatectomia , Regeneração Hepática/fisiologia , Fígado/cirurgia , MicroRNAs/fisiologia , Análise em Microsséries , Animais , Biologia Computacional , Fator de Crescimento de Hepatócito/fisiologia , Interleucina-6/fisiologia , Fígado/fisiologia , Masculino , Modelos Animais , Antígeno Nuclear de Célula em Proliferação/fisiologia , Ratos , Ratos Wistar , Fatores de Tempo
12.
Infect Immun ; 79(2): 928-36, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21149592

RESUMO

Thrombopoietin (Tpo), which primarily regulates megakaryopoiesis, and its receptor (c-Mpl) are expressed in the brain, where Tpo exhibits proapototic effects on neurons. In the present study, we investigated the implication of Tpo in experimental pneumococcal meningitis. Following intrathecal infection with the encapsulated Streptococcus pneumoniae strain D39, we observed upregulation of Tpo mRNA expression at 12 h and 24 h in brain homogenates of wild-type C57BL/6 mice. c-Mpl mRNA expression was upregulated at 12 h and returned to baseline at 24 h. Compared to wild-type mice, mutants with homozygous Tpo receptor ablation (c-Mpl(-/-)) displayed reduced microglial activation and neuronal apoptosis in the dentate gyrus. Concentrations of bacteria in blood or cerebrospinal fluid (CSF), as well as CSF pleocytosis, were not significantly different between wild-type and c-Mpl(-/-) mice. In human postmortem brain, Tpo protein was colocalized to macrophages during encephalitis. In murine primary microglia and RAW264.7 macrophages, upregulation of Tpo mRNA was induced by D39-conditioned medium but not by bacterial lipopeptide or by medium conditioned by pneumococcal mutants defective in hydrogen peroxide formation (ΔspxB) or pneumolysin (Δpln). We conclude that Tpo acts as a mediator of neuronal damage in bacterial meningitis.


Assuntos
Meningite Pneumocócica/patologia , Neurônios/patologia , Trombopoetina/metabolismo , Animais , Anticorpos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Streptococcus pneumoniae , Trombopoetina/genética
13.
PLoS One ; 5(8): e12038, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20706634

RESUMO

BACKGROUND: Reduced chemosensitivity of solid cancer cells represents a pivotal obstacle in clinical oncology. Hence, the molecular characterization of pathways regulating chemosensitivity is a central prerequisite to improve cancer therapy. The hypoxia-inducible factor HIF-1alpha has been linked to chemosensitivity while the underlying molecular mechanisms remain largely elusive. Therefore, we comprehensively analysed HIF-1alpha's role in determining chemosensitivity focussing on responsible molecular pathways. METHODOLOGY AND PRINCIPAL FINDINGS: RNA interference was applied to inactivate HIF-1alpha or p53 in the human gastric cancer cell lines AGS and MKN28. The chemotherapeutic agents 5-fluorouracil and cisplatin were used and chemosensitivity was assessed by cell proliferation assays as well as determination of cell cycle distribution and apoptosis. Expression of p53 and p53 target proteins was analyzed by western blot. NF-kappaB activity was characterized by means of electrophoretic mobility shift assay. Inactivation of HIF-1alpha in gastric cancer cells resulted in robust elevation of chemosensitivity. Accordingly, HIF-1alpha-competent cells displayed a significant reduction of chemotherapy-induced senescence and apoptosis. Remarkably, this phenotype was completely absent in p53 mutant cells while inactivation of p53 per se did not affect chemosensitivity. HIF-1alpha markedly suppressed chemotherapy-induced activation of p53 and p21 as well as the retinoblastoma protein, eventually resulting in cell cycle arrest. Reduced formation of reactive oxygen species in HIF-1alpha-competent cells was identified as the molecular mechanism of HIF-1alpha-mediated inhibition of p53. Furthermore, loss of HIF-1alpha abrogated, in a p53-dependent manner, chemotherapy-induced DNA-binding of NF-kappaB and expression of anti-apoptotic NF-kappaB target genes. Accordingly, reconstitution of the NF-kappaB subunit p65 reversed the increased chemosensitivity of HIF-1alpha-deficient cells. CONCLUSION AND SIGNIFICANCE: In summary, we identified HIF-1alpha as a potent regulator of p53 and NF-kappaB activity under conditions of genotoxic stress. We conclude that p53 mutations in human tumors hold the potential to confound the efficacy of HIF-1-inhibitors in cancer therapy.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Gástricas/metabolismo , Fator de Transcrição RelA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Técnicas de Silenciamento de Genes , Humanos , Mutação , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
14.
BMC Dev Biol ; 10: 77, 2010 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-20667107

RESUMO

BACKGROUND: Thrombopoietin (Thpo) and its receptor (Mpl), which regulate megakaryopoiesis, are expressed in the central nervous system (CNS), where Thpo is thought to exert pro-apoptotic effects on newly generated neurons. Mpl expression has been analysed in brain tissue on transcript level and in cultured primary rat neurons and astrocytes on protein level. Herein, we analysed Mpl expression in the developing and adult murine CNS by immunohistochemistry and investigated the brain of mice with homozygous Mpl deficiency (Mpl-/-) by MRI. RESULTS: Mpl was not detectable at developmental stages E12 to E15 in any resident cells of the CNS. From E18 onwards, robust Mpl expression was found in various brain areas, including cerebral cortex, olfactory bulb, thalamus, hypothalamus, medulla, pons, and the grey matter of spinal cord. However, major developmental changes became obvious: In the subventricular zone of the cerebral cortex Mpl expression occurred only during late gestation, while in the hippocampus Mpl expression was detectable for first time at stage P4. In the white matter of the cerebellum Mpl expression was restricted to the perinatal period. In the adult cerebellum, Mpl expression switched to Purkinje cell. The majority of other Mpl-positive cells were NeuN-positive neurons. None of the cells could be double-labelled with astrocyte marker GFAP. Mpl-/- mice showed no gross abnormalities of the brain. CONCLUSIONS: Our data locate Mpl expression to neurons at different subdivisions of the spinal cord, rhombencephalon, midbrain and prosencephalon. Besides neuronal cells Mpl protein is also expressed in Purkinje cells of the adult cerebellum.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Expressão Gênica , Receptores de Trombopoetina/genética , Animais , Encéfalo/citologia , Células Cultivadas , Camundongos , Ratos , Trombopoetina/metabolismo
15.
Blood ; 116(11): 1867-75, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20538807

RESUMO

Long-lived plasma cells in the bone marrow produce memory antibodies that provide immune protection persisting for decades after infection or vaccination but can also contribute to autoimmune and allergic diseases. However, the composition of the microenvironmental niches that are important for the generation and maintenance of these cells is only poorly understood. Here, we demonstrate that, within the bone marrow, plasma cells interact with the platelet precursors (megakaryocytes), which produce the prominent plasma cell survival factors APRIL (a proliferation-inducing ligand) and IL-6 (interleukin-6). Accordingly, reduced numbers of immature and mature plasma cells are found in the bone marrow of mice deficient for the thrombopoietin receptor (c-mpl) that show impaired megakaryopoiesis. After immunization, accumulation of antigen-specific plasma cells in the bone marrow is disturbed in these mice. Vice versa, injection of thrombopoietin allows the accumulation and persistence of a larger number of plasma cells generated in the course of a specific immune response in wild-type mice. These results demonstrate that megakaryocytes constitute an important component of the niche for long-lived plasma cells in the bone marrow.


Assuntos
Células da Medula Óssea/metabolismo , Megacariócitos/metabolismo , Plasmócitos/metabolismo , Nicho de Células-Tronco/metabolismo , Animais , Células da Medula Óssea/citologia , Comunicação Celular/efeitos dos fármacos , Contagem de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Ovalbumina/imunologia , Ovalbumina/farmacologia , Plasmócitos/citologia , Plasmócitos/efeitos dos fármacos , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/citologia , Trombopoetina/farmacologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
16.
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
17.
J Interferon Cytokine Res ; 30(2): 55-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20028206

RESUMO

The clinical prognosis of children with high-stage neuroblastoma is still poor. Therapeutic approaches include surgery and cellular differentiation by retinoic acid, but also experimental interleukin-based immune modulation. However, the molecular mechanisms of all-trans retinoic acid (ATRA)-induced differentiation of neuroblastoma cells are incompletely understood. Herein, we examined the effect of ATRA on the activity of the interleukin-18 (IL-18) system in human SH-SY5Y neuroblastoma cells. It is shown that SH-SY5Y cells express IL-18 receptor (IL-18R) and the secreted antagonist IL-18-binding protein (IL-18BP), but no IL-18. SH-SY5Y cells are highly sensitive to ATRA treatment and react by cellular differentiation from a neuroblastic toward a more neuronal phenotype. This was associated with induction of IL-18 and reduction of IL-18BP expression, while IL-18R expression remained stable. Thereby, we identified the IL-18 system as a novel target of ATRA in neuroblastoma cells that might contribute to the therapeutic properties of retinoids in treatment of neuroblastoma.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-18/imunologia , Neuroblastoma/imunologia , Neuroblastoma/patologia , Receptores de Interleucina-18/biossíntese , Tretinoína/farmacologia , Linhagem Celular Tumoral , Humanos
18.
J Mol Endocrinol ; 43(4): 157-69, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19491195

RESUMO

Homozygous deficiency of the transcription factor Gata4 in mice causes lethality due to defects in ventral morphogenesis and heart tube formation. There is increasing evidence demonstrating that GATA4 function is also relevant for normal developed organ systems, including the heart and endocrinum. To analyze the implication of Gata4 beyond development, we generated transgenic mice expressing inducible small interfering RNA against Gata4. In longitudinal analysis, efficient suppression of Gata4 mRNA (down to 80% of wild-type levels) and protein expression in the heart was detected 38 days after induction of Gata4 short hairpin RNA. Decreased Gata4 expression was associated with reduction in the expression of known cardiac target genes, but the function of the heart remained unperturbed at 20-30% of normal Gata4 levels. Interestingly, Gata4 expression was almost abolished in the ovary and testis. This was accompanied in the testis by a significant reduction of GATA4 downstream target genes, such as the genes encoding Mullerian inhibiting substance (MIS) and steroidogenic acute regulatory (StAR) protein. By contrast, expression levels of Mis and Star were only slightly modified in the ovary, and concentrations of circulating FSH and LH were normal in female transgenic mice after induction of Gata4 short hairpin RNA. However, inhibition of Gata4 expression led to the formation of ovarian teratoma in 10% of females. Histology of the teratomas showed predominantly ectodermal and mesodermal structures. Our data demonstrate that Gata4 is critically involved in the function and integrity of the gonads in vivo.


Assuntos
Fator de Transcrição GATA4/fisiologia , Gônadas/metabolismo , Miocárdio/metabolismo , RNA Interferente Pequeno/fisiologia , Animais , Southern Blotting , Western Blotting , Linhagem Celular , Feminino , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Reação em Cadeia da Polimerase , Interferência de RNA , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Pediatrics ; 122(2): 375-82, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18676556

RESUMO

OBJECTIVE: Erythropoietin has been shown to be protective against hypoxic-ischemic and inflammatory injuries in cell culture, animal models of brain injury, and clinical trials of adult humans. The rationale for our study was that early administration of high-dose recombinant human erythropoietin may reduce perinatal brain injury (intraventricular hemorrhage and periventricular leukomalacia) in very preterm infants and improve neurodevelopmental outcome. We investigated whether administration of high-dose recombinant human erythropoietin to very preterm infants shortly after birth and subsequently during the first 2 days is safe in terms of short-term outcome. METHODS: This was a randomized, double-masked, single-center trial with a 2:1 allocation in favor of recombinant human erythropoietin. Preterm infants (gestational age: 24 to 31 weeks) were given recombinant human erythropoietin or NaCl 0.9% intravenously 3, 12 to 18, and 36 to 42 hours after birth. RESULTS: The percentage of infants who survived without brain injury or retinopathy was 53% in the recombinant human erythropoietin group and 60% in the placebo group. There were no relevant differences regarding short-term outcomes such as intraventricular hemorrhage, retinopathy, sepsis, necrotizing enterocolitis, and bronchopulmonary dysplasia. For 5 infants who were in the recombinant human erythropoietin group and had a gestational age of <26 weeks, withdrawal of intensive care was decided (3 of 5 with severe bilateral intraventricular hemorrhage, 2 of 5 with pulmonary insufficiency); no infant of the control group died. Recombinant human erythropoietin treatment did not result in significant differences in blood pressure, cerebral oxygenation, hemoglobin, leukocyte, and platelet count. CONCLUSIONS: No significant adverse effects of early high-dose recombinant human erythropoietin treatment in very preterm infants were identified. These results enable us to embark on a large multicenter trial with the aim of determining whether early high-dose administration of recombinant human erythropoietin to very preterm infants improves neurodevelopmental outcome at 24 months' and 5 years' corrected age.


Assuntos
Encefalopatias/prevenção & controle , Deficiências do Desenvolvimento/prevenção & controle , Eritropoetina/administração & dosagem , Doenças do Prematuro/tratamento farmacológico , Recém-Nascido de muito Baixo Peso , Índice de Apgar , Encefalopatias/tratamento farmacológico , Encefalopatias/mortalidade , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/mortalidade , Hemorragia Cerebral/prevenção & controle , Deficiências do Desenvolvimento/tratamento farmacológico , Deficiências do Desenvolvimento/mortalidade , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Seguimentos , Idade Gestacional , Humanos , Recém-Nascido , Doenças do Prematuro/diagnóstico , Doenças do Prematuro/mortalidade , Unidades de Terapia Intensiva Neonatal , Masculino , Dose Máxima Tolerável , Probabilidade , Proteínas Recombinantes , Valores de Referência , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/prevenção & controle , Medição de Risco , Análise de Sobrevida , Resultado do Tratamento
20.
J Pediatr ; 152(5): 728-30, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18410782

RESUMO

Hematopoietic and non-hematopoietic effects of recombinant erythropoietin (Epo) given to preterm infants are controversially discussed. Because renal loss of Epo was significantly higher after intravenous versus subcutaneous Epoetin-beta administration, we suggest a reconsideration of whether subcutaneous recombinant Epo is more efficient and safer because of lower peaks of circulating Epo.


Assuntos
Anemia Neonatal/tratamento farmacológico , Anemia Neonatal/urina , Eritropoetina/administração & dosagem , Eritropoetina/farmacocinética , Doenças do Prematuro/tratamento farmacológico , Doenças do Prematuro/urina , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Recém-Nascido de muito Baixo Peso , Injeções Intravenosas , Injeções Subcutâneas , Estudos Longitudinais , Proteínas Recombinantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA