Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Am J Hematol ; 99(7): 1326-1337, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38646919

RESUMO

Emerging evidence shows the crucial role of inflammation (particularly NF-κB pathway) in the development and progression of myelofibrosis (MF), becoming a promising therapeutic target. Furthermore, tailoring treatment with currently available JAK inhibitors (such as ruxolitinib or fedratinib) does not modify the natural history of the disease and has important limitations, including cytopenias. Since recent studies have highlighted the role of miR-146a, a negative regulator of the NF-κB pathway, in the pathogenesis of MF; here we used miR-146a-/- (KO) mice, a MF-like model lacking driver mutations, to investigate whether pharmacological inhibition of JAK/STAT and/or NF-κB pathways may reverse the myelofibrotic phenotype of these mice. Specifically, we tested the JAK1/2 inhibitor, ruxolitinib; the NF-κB inhibitor via IKKα/ß, BMS-345541; both inhibitors in combination; or a dual inhibitor of both pathways (JAK2/IRAK1), pacritinib. Although all treatments decreased spleen size and partially recovered its architecture, only NF-κB inhibition, either using BMS-345541 (alone or in combination) or pacritinib, resulted in a reduction of extramedullary hematopoiesis, bone marrow (BM) fibrosis and osteosclerosis, along with an attenuation of the exacerbated inflammatory state (via IL-1ß and TNFα). However, although dual inhibitor improved anemia and reversed thrombocytopenia, the combined therapy worsened anemia by inducing BM hypoplasia. Both therapeutic options reduced NF-κB and JAK/STAT signaling in a context of JAK2V617F-driven clonal hematopoiesis. Additionally, combined treatment reduced both COL1A1 and IL-6 production in an in vitro model mimicking JAK2-driven fibrosis. In conclusion, NF-κB inhibition reduces, in vitro and in vivo, disease burden and BM fibrosis, which could provide benefits in myelofibrosis patients.


Assuntos
Camundongos Knockout , MicroRNAs , NF-kappa B , Nitrilas , Mielofibrose Primária , Pirazóis , Pirimidinas , Animais , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/genética , MicroRNAs/genética , Camundongos , NF-kappa B/metabolismo , Nitrilas/uso terapêutico , Nitrilas/farmacologia , Pirimidinas/uso terapêutico , Pirimidinas/farmacologia , Pirazóis/uso terapêutico , Pirazóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Modelos Animais de Doenças , Hematopoese Extramedular/efeitos dos fármacos
3.
JCI Insight ; 6(6)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33600377

RESUMO

The splenic microenvironment regulates hematopoietic stem and progenitor cell (HSPC) function, particularly during demand-adapted hematopoiesis; however, practical strategies to enhance splenic support of transplanted HSPCs have proved elusive. We have previously demonstrated that inhibiting 15-hydroxyprostaglandin dehydrogenase (15-PGDH), using the small molecule (+)SW033291 (PGDHi), increases BM prostaglandin E2 (PGE2) levels, expands HSPC numbers, and accelerates hematologic reconstitution after BM transplantation (BMT) in mice. Here we demonstrate that the splenic microenvironment, specifically 15-PGDH high-expressing macrophages, megakaryocytes (MKs), and mast cells (MCs), regulates steady-state hematopoiesis and potentiates recovery after BMT. Notably, PGDHi-induced neutrophil, platelet, and HSPC recovery were highly attenuated in splenectomized mice. PGDHi induced nonpathologic splenic extramedullary hematopoiesis at steady state, and pretransplant PGDHi enhanced the homing of transplanted cells to the spleen. 15-PGDH enzymatic activity localized specifically to macrophages, MK lineage cells, and MCs, identifying these cell types as likely coordinating the impact of PGDHi on splenic HSPCs. These findings suggest that 15-PGDH expression marks HSC niche cell types that regulate hematopoietic regeneration. Therefore, PGDHi provides a well-tolerated strategy to therapeutically target multiple HSC niches, promote hematopoietic regeneration, and improve clinical outcomes of BMT.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hematopoese Extramedular/efeitos dos fármacos , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Regeneração , Baço/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Baço/enzimologia , Baço/metabolismo
4.
Int J Hematol ; 113(3): 348-361, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33398631

RESUMO

Histidine decarboxylase (HDC), a histamine synthase, is expressed in various hematopoietic cells and is induced by hematopoietic cytokines such as granulocyte colony-stimulating factor (G-CSF). We previously showed that nitrogen-containing bisphosphonate (NBP)-treatment induces extramedullary hematopoiesis via G-CSF stimulation. However, the function of HDC in NBP-induced medullary and extramedullary hematopoiesis remains unclear. Here, we investigated changes in hematopoiesis in wild-type and HDC-deficient (HDC-KO) mice. NBP treatment did not induce anemia in wild-type or HDC-KO mice, but did produce a gradual increase in serum G-CSF levels in wild-type mice. NBP treatment also enhanced Hdc mRNA expression and erythropoiesis in the spleen and reduced erythropoiesis in bone marrow and the number of vascular adhesion molecule 1 (VCAM-1)-positive macrophages in wild-type mice, as well as increased the levels of hematopoietic progenitor cells and proliferating cells in the spleen and enhanced expression of bone morphogenetic protein 4 (Bmp4), CXC chemokine ligand 12 (Cxcl12), and hypoxia inducible factor 1 (Hif1) in the spleen. However, such changes were not observed in HDC-KO mice. These results suggest that histamine may affect hematopoietic microenvironments of the bone marrow and spleen by changing hematopoiesis-related factors in NBP-induced extramedullary hematopoiesis.


Assuntos
Alendronato/antagonistas & inibidores , Medula Óssea/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Hematopoese Extramedular/efeitos dos fármacos , Histidina Descarboxilase/deficiência , Baço/efeitos dos fármacos , Alendronato/farmacologia , Alendronato/toxicidade , Anemia/induzido quimicamente , Animais , Medula Óssea/metabolismo , Proteína Morfogenética Óssea 4/biossíntese , Proteína Morfogenética Óssea 4/genética , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/genética , Indução Enzimática/efeitos dos fármacos , Células Eritroides/patologia , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos/sangue , Histamina/biossíntese , Histidina Descarboxilase/biossíntese , Histidina Descarboxilase/genética , Histidina Descarboxilase/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Baço/metabolismo
5.
Front Immunol ; 11: 1627, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849551

RESUMO

Dendritic cells (DC) play a key role in the adaptive immune response due to their ability to present antigens and stimulate naïve T cells. Many bacteria and viruses can efficiently target DC, resulting in impairment of their immunostimulatory function or elimination. Hence, the DC compartment requires replenishment following infection to ensure continued operational readiness of the adaptive immune system. Here, we investigated the molecular and cellular mechanisms of inflammation-induced DC generation. We found that infection with viral and bacterial pathogens as well as Toll-like receptor 9 (TLR9) ligation with CpG-oligodeoxynucleotide (CpG-ODN) expanded an erythropoietin (EPO)-dependent TER119+CD11a+ cell population in the spleen that had the capacity to differentiate into TER119+CD11chigh and TER119-CD11chigh cells both in vitro and in vivo. TER119+CD11chigh cells contributed to the conventional DC pool in the spleen and specifically increased in lymph nodes draining the site of local inflammation. Our results reveal a so far undescribed inflammatory EPO-dependent pathway of DC differentiation and establish a mechanistic link between innate immune recognition of potential immunosuppressive pathogens and the maintenance of the DC pool during and after infection.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Eritropoetina/metabolismo , Imunidade Inata , Infecções/etiologia , Infecções/metabolismo , Animais , Biomarcadores , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Antígeno CD11c/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Eritropoetina/farmacologia , Feminino , Hematopoese Extramedular/efeitos dos fármacos , Hematopoese Extramedular/imunologia , Imunofenotipagem , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Transgênicos , Oligodesoxirribonucleotídeos/farmacologia , Baço/imunologia , Baço/metabolismo , Baço/patologia
6.
Clin Nucl Med ; 45(8): e373-e375, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32558708

RESUMO

We report the case of a 23-year-old man with nodal EMH (extramedullary hematopoiesis) occurring during treatment for a stage IIA "gray-zone" lymphoma. Although it is often related to myeloproliferative bone marrow disease, benign etiologies such as lenograstim treatment after chemotherapy can also induce EMH and be responsible for false-positive F-FDG PET/CT examinations. In this respect, GLUT overexpression in hematopoietic lineages and macrophages of the inflammatory environment are responsible for increased F-FDG uptake. Histopathologic confirmation of new hypermetabolic lesions on follow-up PET/CT may be required when the new lesions do not conform with the treatment responses in the preexisting lesions.


Assuntos
Fluordesoxiglucose F18 , Hematopoese Extramedular/efeitos dos fármacos , Lenograstim/efeitos adversos , Linfoma/tratamento farmacológico , Linfoma/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Humanos , Linfoma/diagnóstico por imagem , Masculino , Estadiamento de Neoplasias , Adulto Jovem
7.
Sci Rep ; 8(1): 8308, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844356

RESUMO

Extramedullary hematopoiesis (EMH) in postnatal life is a pathological process in which the differentiation of hematopoietic stem/progenitor cells (HSPCs) occurs outside the bone marrow (BM) to respond to hematopoietic emergencies. The spleen is a major site for EMH; however, the cellular and molecular nature of the stromal cell components supporting HSPC maintenance, the niche for EMH in the spleen remain poorly understood compared to the growing understanding of the BM niche at the steady-state as well as in emergency hematopoiesis. In the present study, we demonstrate that mesenchymal progenitor-like cells expressing Tlx1, an essential transcription factor for spleen organogenesis, and selectively localized in the perifollicular region of the red pulp of the spleen, are a major source of HSPC niche factors. Consistently, overexpression of Tlx1 in situ induces EMH, which is associated with mobilization of HSPC into the circulation and their recruitment into the spleen where they proliferate and differentiate. The alterations in the splenic microenvironment induced by Tlx1 overexpression in situ phenocopy lipopolysaccharide (LPS)-induced EMH, and the conditional loss of Tlx1 abolished LPS-induced splenic EMH. These findings indicate that activation of Tlx1 expression in the postnatal splenic mesenchymal cells is critical for the development of splenic EMH.


Assuntos
Hematopoese Extramedular/fisiologia , Proteínas de Homeodomínio/fisiologia , Baço/citologia , Nicho de Células-Tronco , Animais , Proliferação de Células , Hematopoese Extramedular/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout
8.
Int J Mol Sci ; 19(3)2018 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-29562644

RESUMO

Myelofibrosis (MF) is a clinical manifestation of chronic BCR-ABL1-negative chronic myeloproliferative neoplasms. Splenomegaly is one of the major clinical manifestations of MF and is directly linked to splenic extramedullary hematopoiesis (EMH). EMH is associated with abnormal trafficking patterns of clonal hematopoietic cells due to the dysregulated bone marrow (BM) microenvironment leading to progressive splenomegaly. Several recent data have emphasized the role of several cytokines for splenic EMH. Alteration of CXCL12/CXCR4 pathway could also lead to splenic EMH by migrated clonal hematopoietic cells from BM to the spleen. Moreover, low Gata1 expression was found to be significantly associated with the EMH. Several gene mutations were found to be associated with significant splenomegaly in MF. In recent data, JAK2V617F homozygous mutation was associated with a larger spleen size. In other data, CALR mutations in MF were signigicantly associated with longer larger splenomegaly-free survivals than others. In addition, MF patients with ≥1 mutations in AZXL1, EZH1 or IDH1/2 had significantly low spleen reduction response in ruxolitinib treatment. Developments of JAK inhibitors, such as ruxolitinib, pacritinib, momelotinib, and febratinib enabled the effective management in MF patients. Especially, significant spleen reduction responses of the drugs were demonstrated in several randomized clinical studies, although those could not eradicate allele burdens of MF.


Assuntos
Hematopoese Extramedular/fisiologia , Janus Quinases/genética , Mielofibrose Primária/genética , Mielofibrose Primária/patologia , Esplenomegalia/genética , Medula Óssea/patologia , Movimento Celular , Quimiocina CXCL12/metabolismo , Hematopoese Extramedular/efeitos dos fármacos , Humanos , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Mutação , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/fisiopatologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores CXCR4/metabolismo , Baço/patologia , Esplenomegalia/tratamento farmacológico , Esplenomegalia/fisiopatologia
9.
Pediatr Blood Cancer ; 65(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29049846

RESUMO

The congenital dyserythropoietic anemias (CDAs) are a group of rare inherited blood disorders characterized by ineffective erythropoiesis as the principal cause of anemia. We present a child with CDA 1b-the rarest and least well-described type-due to a mutation in the C15orf41 gene. The patient presented with severe in utero and neonatal manifestations, typical peripheral limb anomalies as well as rarely reported cardiac manifestations, visual impairment, short stature, and hip dysplasia. Anemia was complicated by iron overload and pronounced extra medullary erythropoiesis leading to skull deformities. The patient responded to treatment with pegylated interferon alfa-2a.


Assuntos
Anemia Diseritropoética Congênita , Hematopoese Extramedular/efeitos dos fármacos , Interferon alfa-2/administração & dosagem , Sobrecarga de Ferro , Mutação , Anemia Diseritropoética Congênita/diagnóstico por imagem , Anemia Diseritropoética Congênita/tratamento farmacológico , Anemia Diseritropoética Congênita/genética , Humanos , Recém-Nascido , Sobrecarga de Ferro/diagnóstico por imagem , Sobrecarga de Ferro/tratamento farmacológico , Sobrecarga de Ferro/genética , Masculino , Crânio/anormalidades , Crânio/diagnóstico por imagem
10.
J Clin Invest ; 127(9): 3392-3401, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28783041

RESUMO

Extramedullary hematopoiesis (EMH) is induced during pregnancy to support rapid expansion of maternal blood volume. EMH activation requires hematopoietic stem cell (HSC) proliferation and mobilization, processes that depend upon estrogen receptor α (ERα) in HSCs. Here we show that treating mice with estradiol to model estradiol increases during pregnancy induced HSC proliferation in the bone marrow but not HSC mobilization. Treatment with the alternative ERα ligand 27-hydroxycholesterol (27HC) induced ERα-dependent HSC mobilization and EMH but not HSC division in the bone marrow. During pregnancy, 27HC levels increased in hematopoietic stem/progenitor cells as a result of CYP27A1, a cholesterol hydroxylase. Cyp27a1-deficient mice had significantly reduced 27HC levels, HSC mobilization, and EMH during pregnancy but normal bone marrow hematopoiesis and EMH in response to bleeding or G-CSF treatment. Distinct hematopoietic stresses thus induce EMH through different mechanisms. Two different ERα ligands, estradiol and 27HC, work together to promote EMH during pregnancy, revealing a collaboration of hormonal and metabolic mechanisms as well as a physiological function for 27HC in normal mice.


Assuntos
Hematopoese Extramedular/efeitos dos fármacos , Mobilização de Células-Tronco Hematopoéticas/métodos , Hidroxicolesteróis/farmacologia , Animais , Células da Medula Óssea/citologia , Proliferação de Células , Colestanotriol 26-Mono-Oxigenase/genética , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/fisiologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Prenhez , Células-Tronco/citologia
11.
Intern Med ; 56(10): 1213-1217, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28502939

RESUMO

A 64-year-old woman was diagnosed to have refractory cytopenia with multilineage dysplasia (RCMD) including an increased number of sideroblasts in the bone marrow (BM). Computed tomography (CT) revealed a presacral mass which showed iso- or high-intensity signals according to T1-weighted and hypo-intensity signals on T2-weighted magnetic resonance imaging (MRI). CT-guided biopsy revealed the presence of hematopoietic tissue with features that correlated with the BM findings. While the formation of extramedullary hematopoiesis in the presacral area is rare, it is important to differentiate it from other parasacral tumors even though such differentiation is often difficult. This patient demonstrated atypical MRI signals possibly due to an increase in the cellular iron content of the erythroid precursors.


Assuntos
Medula Óssea/efeitos dos fármacos , Medula Óssea/fisiopatologia , Neoplasias Ósseas/complicações , Hematopoese Extramedular/efeitos dos fármacos , Ferro/sangue , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/tratamento farmacológico , Medula Óssea/diagnóstico por imagem , Neoplasias Ósseas/diagnóstico por imagem , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/diagnóstico por imagem , Síndromes Mielodisplásicas/etiologia , Tomografia Computadorizada por Raios X/métodos
12.
J Appl Toxicol ; 37(8): 913-921, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28138994

RESUMO

Hexahydro-1-nitroso-3,5-dinitro-1,3,5-triazine (MNX), environmental degradation product of munitions hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX), causes seizures in rats with acute oral exposure like parent RDX. Our previous studies have additionally reported hematotoxicity with acute MNX exposure manifested as myelosuppression, anemia and splenic hemosiderosis. This study explored whether MNX administered subchronically continued to target bone marrow to elicit peripheral blood cytopenia. Female Sprague-Dawley rats were gavaged daily for 4 or 6 weeks with 47 mg kg-1 day-1 MNX (» LD50 ) or vehicle (5% dimethyl sulfoxide in corn oil) and hematological and clinical chemistry parameters, spleen weights, spleen and bone marrow histopathology and immunohistochemistry with ED1 anti-CD68 macrophage marker were evaluated 24 h after the last dose. Unexpectedly, no decrease in blood erythroid parameters was seen with subchronic MNX and convulsions and tremors ceased after 2 weeks of treatment. Toxicological effects observed were MNX-induced increases in blood granulocyte and platelet counts and in bone marrow megakaryocyte and ED1+ -macrophage density. MNX was without effect on bone marrow cellularity and picrosirius red stained/collagen fiber deposition. Spleen weight increased modestly with extramedullary hematopoiesis evident, but hemosiderin and relative red and white pulp areas were unaffected. Collectively, this study demonstrated that erythroid effects characteristic of acute MNX exposure were not evident with subchronic exposure. However, megakaryocyte proliferation in bone marrow coincident with thrombocytosis after subchronic MNX exposure suggested continued hematotoxicity, but with a qualitatively different outcome. Granulocytosis and increased bone marrow macrophages implicated an inflammatory component in MNX hematotoxicity. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Medula Óssea/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Substâncias Explosivas/toxicidade , Macrófagos/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Triazinas/toxicidade , Animais , Biodegradação Ambiental , Contagem de Células Sanguíneas , Medula Óssea/imunologia , Medula Óssea/patologia , Relação Dose-Resposta a Droga , Feminino , Hematopoese Extramedular/efeitos dos fármacos , Macrófagos/imunologia , Megacariócitos/citologia , Tamanho do Órgão/efeitos dos fármacos , Ratos Sprague-Dawley , Baço/efeitos dos fármacos , Baço/patologia
13.
Oncogene ; 36(26): 3760-3771, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28218904

RESUMO

FTY720 (also called fingolimod) is recognized as an immunosuppressant and has been approved by the Food and Drug Administration to treat refractory multiple sclerosis. However, long-term administration of FTY720 potentially increases the risk for cancer in recipients. The underlying mechanisms remain poorly understood. Herein, we provided evidence that FTY720 administration potentiated tumor growth. Mechanistically, FTY720 enhanced extramedullary hematopoiesis and massive accumulation of myeloid-derived suppressor cells (MDSCs), which actively suppressed antitumor immune responses. Granulocyte-macrophage colony-stimulating factor (GM-CSF), mainly produced by MDSCs, was identified as a key factor to mediate these effects of FTY720 in tumor microenvironment. Furthermore, we showed that FTY720 triggers MDSCs to release GM-CSF via S1P receptor 3 (S1pr3) through Rho kinase and extracellular signal-regulated kinase-dependent pathway. Thus, our findings provide mechanistic explanation for the protumorigenic potentials of FTY720 and suggest that targeting S1pr3 simultaneously may be beneficial for the patients receiving FTY720 treatment.


Assuntos
Cloridrato de Fingolimode/efeitos adversos , Hematopoese Extramedular/efeitos dos fármacos , Imunossupressores/efeitos adversos , Células Mieloides/efeitos dos fármacos , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Cloridrato de Fingolimode/farmacologia , Humanos , Imunossupressores/farmacologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Mieloides/citologia , Células Mieloides/imunologia , Microambiente Tumoral/efeitos dos fármacos
15.
Stem Cells ; 34(12): 2902-2915, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27422171

RESUMO

Impaired T lymphopoiesis is associated with immunosuppression of the adaptive immune response and plays a role in the morbidity and mortality of patients and animal models of sepsis. Although previous studies examined several intrathymic mechanisms that negatively affect T lymphopoiesis, the extrathymic mechanisms remain poorly understood. Here, we report a dramatic decrease in the percentage of early T lineage progenitors (ETPs) in three models of sepsis in mice (cecal ligation and puncture, lipopolysaccharide continuous injection, and poly I:C continuous injection). However, septic mice did not show a decrease in the number of bone marrow (BM) precursor cells. Instead, the BM progenitors for ETPs expressed reduced mRNA levels of CC chemokine receptor (CCR) 7, CCR9 and P-selectin glycoprotein ligand 1, and exhibited impaired homing capacity in vitro and in vivo. Furthermore, RNA-Seq analysis and real-time PCR showed a marked downregulation of several lymphoid-related genes in hematopoietic stem and progenitor cells. Hematopoietic stem and progenitor cells differentiated into myeloid cells but failed to generate T lymphocytes in vitro and in vivo. Our results indicate that the depletion of ETPs in septic mice might be a consequence of an impaired migration of BM progenitors to the thymus, as well as a defect in lymphoid lineage commitment. Stem Cells 2016;34:2902-2915.


Assuntos
Linfopoese , Sepse/complicações , Timo/patologia , Animais , Atrofia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Perfilação da Expressão Gênica , Hematopoese Extramedular/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Contagem de Linfócitos , Linfopoese/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Mielopoese/efeitos dos fármacos , Poli I-C/farmacologia , Receptores de Quimiocinas/metabolismo , Sepse/genética , Sepse/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Timo/efeitos dos fármacos , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
17.
J Cell Mol Med ; 19(11): 2575-86, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26153045

RESUMO

Cyclophosphamide (CY) is a chemotherapeutic agent used for cancer and immunological diseases. It induces cytotoxicity of bone marrow and causes myelosuppression and extramedullary haematopoiesis (EMH) in treated patients. EMH is characterized with the emergence of multipotent haematopoietic progenitors most likely in the spleen and liver. Previous studies indicated that a Chinese medicine, ginsenoside Rg1, confers a significant effect to elevate the number of lineage (Lin(-) ) Sca-1(+) c-Kit(+) haematopoietic stem and progenitor cells (HSPCs) and restore the function of bone marrow in CY-treated myelosuppressed mice. However, whether the amelioration of bone marrow by Rg1 accompanies an alleviation of EMH in the spleen was still unknown. In our study, the cellularity and weight of the spleen were significantly reduced after Rg1 treatment in CY-treated mice. Moreover, the number of c-Kit(+) HSPCs was significantly decreased but not as a result of apoptosis, indicating that Rg1 alleviated EMH of the spleen induced by CY. Unexpectedly, the proliferation activity of c-Kit(+) HSPCs was only up-regulated in the spleen, but not in the bone marrow, after Rg1 treatment in CY-treated mice. We also found that a fraction of c-Kit(+) /CD45(+) HSPCs was simultaneously increased in the circulation after Rg1 treatment. Interestingly, the effects of Rg1 on the elevation of HSPCs in bone marrow and in the peripheral blood were suppressed in CY-treated splenectomized mice. These results demonstrated that Rg1 improves myelosuppression induced by CY through its action on the proliferation of HSPCs in EMH of the spleen and migration of HSPCs from the spleen to the bone marrow.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Medula Óssea/fisiopatologia , Ciclofosfamida/farmacologia , Ginsenosídeos/administração & dosagem , Hematopoese Extramedular/efeitos dos fármacos , Hematopoese , Baço/fisiopatologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ginsenosídeos/uso terapêutico , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos
18.
Endocrinology ; 156(8): 2821-30, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26061726

RESUMO

The self-renewal and differentiation of hematopoietic stem cells (HSCs) in bone marrow are essential to replenish all blood cell types, but how this process is influenced by diet remains largely unclear. Here we show that a diet rich in fish oils promotes self-renewal of HSCs and extramedullary hematopoiesis. Chronic intake of a fish oil-rich diet increases the abundance of HSCs, alters the hematopoietic microenvironment, and, intriguingly, induces the expression of matrix metalloproteinase 12 (MMP12) in the bone marrow. Pointing to a direct effect of fish oil on MMP12 expression, omega-3 polyunsaturated fatty acids induce the expression of MMP12 in a dose-dependent manner in bone marrow cells. Importantly, down-regulation of MMP12 activity using an MMP12-specific inhibitor attenuates diet-induced myelopoiesis in both bone marrow and spleen. Thus, a fish oil-rich diet promotes hematopoiesis in the bone marrow and spleen, in part via the activity of MMP12. Taken together, these data provide new insights into diet-mediated regulation of hematopoiesis.


Assuntos
Dieta , Óleos de Peixe/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ácidos Graxos Ômega-3/farmacologia , Hematopoese Extramedular/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/efeitos dos fármacos
19.
Hematology ; 20(1): 53-7, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24717020

RESUMO

OBJECTIVE AND IMPORTANCE: Extramedullary hematopoiesis (EMH) is evidenced by erythropoietic masses, which occurs as a compensatory mechanism to overcome hypoxia during chronic anemia. EMH masses in spinal cord could lead to cord compression and neurological symptoms. Besides transfusion, radiotherapy, and surgery, hydroxyurea (HU) is also a treatment strategy in EMH. CLINICAL PRESENTATION: We described four cases of beta thalassemia with EMH who were treated with HU as a monotherapy. INTERVENTION (AND TECHNIQUE): HU therapy was done in all patients without any transfusion during therapy. CONCLUSION: HU is a good treatment option for patients with EMH and it could be a substitute for radiotherapy and invasive surgery or regular blood transfusion.


Assuntos
Antineoplásicos/uso terapêutico , Hematopoese Extramedular/efeitos dos fármacos , Hidroxiureia/uso terapêutico , Talassemia beta/tratamento farmacológico , Adulto , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Adulto Jovem
20.
Inhal Toxicol ; 26(12): 697-707, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25140454

RESUMO

Spot welding is used in the automotive and aircraft industries, where high-speed, repetitive welding is needed to join thin sections of metal. Epoxy adhesives are applied as sealers to the metal seams. Pulmonary function abnormalities and airway irritation have been reported in spot welders, but no animal toxicology studies exist. Therefore, the goal of this study was to investigate vascular, immune and lung toxicity measures after exposure to these metal fumes in an animal model. Male Sprague-Dawley rats were exposed by inhalation to 25 mg/m³ to either mild-steel spot welding aerosols with sparking (high metal, HM) or without sparking (low metal, LM) for 4 h/d for 3, 8 and 13 d. Shams were exposed to filtered air. Bronchoalveolar lavage (BAL), lung gene expression and ex vivo BAL cell challenge were performed to assess lung toxicity. Lung resistance (R(L)) was evaluated before and after challenge with inhaled methacholine (MCh). Functional assessment of the vascular endothelium in isolated rat tail arteries and leukocyte differentiation in the spleen and lymph nodes via flow cytometry was also done. Immediately after exposure, baseline R(L) was significantly elevated in the LM spot welding aerosols, but returned to control level by 24 h postexposure. Airway reactivity to MCh was unaffected. Lung inflammation and cytotoxicity were mild and transient. Lung epithelial permeability was significantly increased after 3 and 8 d, but not after 13 d of exposure to the HM aerosol. HM aerosols also caused vascular endothelial dysfunction and increased CD4+, CD8+ and B cells in the spleen. Only LM aerosols caused increased IL-6 and MCP-1 levels compared with sham after ex vivo LPS stimulation in BAL macrophages. Acute inhalation of mild-steel spot welding fumes at occupationally relevant concentrations may act as an irritant as evidenced by the increased R(L) and result in endothelial dysfunction, but otherwise had minor effects on the lung.


Assuntos
Poluentes Ocupacionais do Ar/toxicidade , Endotélio Vascular/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Pulmão/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Vasculite/induzido quimicamente , Soldagem , Adesivos/química , Aerossóis , Animais , Células Cultivadas , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Incêndios , Hematopoese Extramedular/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Leucócitos/patologia , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Masculino , Ratos Sprague-Dawley , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Organismos Livres de Patógenos Específicos , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia , Aço/química , Testes de Toxicidade Aguda , Vasculite/imunologia , Vasculite/patologia , Vasculite/fisiopatologia , Soldagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA