Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
1.
J Cell Physiol ; 235(10): 6357-6365, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32003015

RESUMO

C-type lectin domain family 11 member A (Clec11a), also known as stem cell growth factor (SCGF), C-type lectin superfamily member 3 (CLECSF3), or osteolectin was initially identified as a growth factor for hematopoietic progenitor cells. The human Clec11a gene encodes a polypeptide of 323 amino acids with characteristics of a secreted glycoprotein encompassing two integrin-binding motifs, RGD (Arg-Gly-Asp) and LDT (Leu-Asp-Thr), a putative leucine zipper domain, and a functional C-type lectin domain. It regulates hematopoietic differentiation and homeostasis and exhibits a protective effect against severe malarial anemia and lipotoxicity. Furthermore, Clec11a promotes the differentiation of mesenchymal progenitors into mature osteoblasts in vitro and plays an important role in the maintenance of adult skeleton age-related bone loss and fracture repair. Receptor ligand binding results in activation of downstream signaling cascades including glycogen synthase kinase 3 (GSK3), ß-catenin, and Wnt, resulting in the expression of osteoblast-related gene transcripts including Alp, Runx2, Lef1, and Axin2. In addition, Clec11a is also associated with the development of several cancers, including leukemia, multiple myeloma, and gastrointestinal tract tumors. To date, however, the mechanisms governing transcription regulation of the Clec11a gene are not known and remain to be uncovered. Understanding the function and mechanism of action of Clec11a will pave the way for the development of Clec11a as a novel therapeutic target for conditions such as cancer, anemia, and skeletal diseases.


Assuntos
Fatores de Crescimento de Células Hematopoéticas/genética , Neoplasias/genética , Sequência de Aminoácidos , Animais , Biologia , Humanos , Estrutura Molecular , Transcrição Gênica/genética
2.
PLoS Biol ; 17(7): e3000350, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31265461

RESUMO

Mutagenic screening is powerful for identifying key genes involved in developmental processes. However, such screens are successful only in lower organisms. Here, we develop a targeted genetic screening approach in mice through combining androgenetic haploid embryonic stem cells (AG-haESCs) and clustered regularly interspaced palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9) technology. We produced a mutant semi-cloned (SC) mice pool by oocyte injection of AG-haESCs carrying constitutively expressed Cas9 and an single guide RNA (sgRNA) library targeting 72 preselected genes in one step and screened for bone-development-related genes through skeletal analysis at birth. This yielded 4 genes: Zic1 and Clec11a, which are required for bone development, and Rln1 and Irx5, which had not been previously considered. Whereas Rln1-/- mice exhibited small skeletal size only at birth, Irx5-/- mice showed skeletal abnormalities both in postnatal and adult phases due to decreased bone mass and increased bone marrow adipogenesis. Mechanistically, iroquois homeobox 5 (IRX5) promotes osteoblastogenesis and inhibits adipogenesis by suppressing peroxisome proliferator activated receptor γ (PPARγ) activation. Thus, AG-haESC-mediated functional mutagenic screening opens new avenues for genetic interrogation of developmental processes in mice.


Assuntos
Desenvolvimento Ósseo/genética , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes/métodos , Testes Genéticos/métodos , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Sistemas CRISPR-Cas , Células Cultivadas , Haploidia , Fatores de Crescimento de Células Hematopoéticas/genética , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Camundongos , Camundongos Knockout , Relaxina/genética , Relaxina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Elife ; 52016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27976999

RESUMO

Bone marrow stromal cells maintain the adult skeleton by forming osteoblasts throughout life that regenerate bone and repair fractures. We discovered that subsets of these stromal cells, osteoblasts, osteocytes, and hypertrophic chondrocytes secrete a C-type lectin domain protein, Clec11a, which promotes osteogenesis. Clec11a-deficient mice appeared developmentally normal and had normal hematopoiesis but reduced limb and vertebral bone. Clec11a-deficient mice exhibited accelerated bone loss during aging, reduced bone strength, and delayed fracture healing. Bone marrow stromal cells from Clec11a-deficient mice showed impaired osteogenic differentiation, but normal adipogenic and chondrogenic differentiation. Recombinant Clec11a promoted osteogenesis by stromal cells in culture and increased bone mass in osteoporotic mice in vivo. Recombinant human Clec11a promoted osteogenesis by human bone marrow stromal cells in culture and in vivo. Clec11a thus maintains the adult skeleton by promoting the differentiation of mesenchymal progenitors into mature osteoblasts. In light of this, we propose to call this factor Osteolectin.


Assuntos
Diferenciação Celular , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Lectinas Tipo C/metabolismo , Células-Tronco Mesenquimais/fisiologia , Osteogênese , Esqueleto/fisiologia , Adulto , Animais , Células Cultivadas , Fatores de Crescimento de Células Hematopoéticas/deficiência , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Lectinas Tipo C/deficiência , Lectinas Tipo C/genética , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
PLoS One ; 11(12): e0167437, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28005985

RESUMO

Psoriasis is a chronic cutaneous inflammatory disease. The immunopathogenesis is a complex interplay between T cells, dendritic cells and the epidermis in which T cells and dendritic cells maintain skin inflammation. Anti-tumour necrosis factor (anti-TNF)-α agents have been approved for therapeutic use across a range of inflammatory disorders including psoriasis, but the anti-inflammatory mechanisms of anti-TNF-α in lesional psoriatic skin are not fully understood. We investigated early events in skin from psoriasis patients after treatment with anti-TNF-α antibodies by use of bioinformatics tools. We used the Human Gene 1.0 ST Array to analyse gene expression in punch biopsies taken from psoriatic patients before and also 4 and 14 days after initiation of treatment with the anti-TNF-α agent adalimumab. The gene expression was analysed by gene set enrichment analysis using the Functional Annotation Tool from DAVID Bioinformatics Resources. The most enriched pathway was visualised by the Pathview Package on Kyoto Encyclopedia of Genes and Genomes (KEGG) graphs. The analysis revealed new very early events in psoriasis after adalimumab treatment. Some of these events have been described after longer periods of anti-TNF-α treatment when clinical and histological changes appear, suggesting that effects of anti-TNF-α treatment on gene expression appear very early before clinical and histological changes. Combining microarray data on biopsies from psoriasis patients with pathway analysis allowed us to integrate in vitro findings into the identification of mechanisms that may be important in vivo. Furthermore, these results may reflect primary effect of anti-TNF-α treatment in contrast to studies of gene expression changes following clinical and histological changes, which may reflect secondary changes correlated to the healing of the skin.


Assuntos
Adalimumab/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Psoríase/tratamento farmacológico , Pele/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Adalimumab/farmacologia , Adulto , Idoso , Anti-Inflamatórios/farmacologia , Citocinas/genética , Citocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Fatores de Crescimento de Células Hematopoéticas/genética , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Humanos , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Análise de Componente Principal , Psoríase/genética , Psoríase/patologia , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Pele/patologia , Fatores de Tempo
5.
Adv Exp Med Biol ; 843: 173-213, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25956299

RESUMO

In the physiological situation, cytokines are pivotal mediators of communication between the maternal tract and the embryo. Compelling evidence shows that cytokines emanating from the oviduct and uterus confer a sophisticated mechanism for 'fine-tuning' of embryo development, influencing a range of cellular events from cell survival and metabolism, through division and differentiation, and potentially exerting long-term impact through epigenetic remodelling. The balance between survival agents, including GM-CSF, CSF1, LIF, HB-EGF and IGFII, against apoptosis-inducing factors such as TNFα, TRAIL and IFNg, influence the course of preimplantation development, causing embryos to develop normally, adapt to varying maternal environments, or in some cases to arrest and undergo demise. Maternal cytokine-mediated pathways help mediate the biological effects of embryo programming, embryo plasticity and adaptation, and maternal tract quality control. Thus maternal cytokines exert influence not only on fertility and pregnancy progression but on the developmental trajectory and health of offspring. Defining a clear understanding of the biology of cytokine networks influencing the embryo is essential to support optimal outcomes in natural and assisted conception.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Blastocisto/metabolismo , Desenvolvimento Embrionário/genética , Tubas Uterinas/metabolismo , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Útero/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose/genética , Blastocisto/citologia , Sobrevivência Celular , Feminino , Fertilização , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Gravidez , Transdução de Sinais
6.
Dev Comp Immunol ; 51(1): 39-47, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25720979

RESUMO

Here we show that knockdown of laminin receptor (Lamr) with PvLamr dsRNA in the whiteleg shrimp Penaeus (Litopenaeus) vannamei (Pv) caused a dramatic reduction specifically in hyaline hemocytes prior to death. Since apoptosis was not detected in hemocytes or hematopoietic cells, other possible causes of hemocyte loss were investigated. Reports that suppression of crustacean hematopoietic factor (CHF)-like protein or hemocyte homeostasis-associated protein (HHAP) also reduced shrimp hemocyte counts led us to carry out yeast two-hybrid (Y2H) and co-immunoprecipitation (co-IP) assays to test for interactions between Lamr and Pv homologues to these proteins (PvCHF-like and PvHHAP). The assays revealed that Lamr bound to both these homologues, but that the homologues did not bind to each other. Subsequent RT-PCR assays confirmed that PvLamr dsRNA injection significantly reduced expression levels for both PvCHF-like and PvHHAP genes. Further work is needed to determine how interaction among these three proteins can regulate shrimp hemocyte homeostasis.


Assuntos
Hemócitos/fisiologia , Penaeidae/imunologia , Receptores de Laminina/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular , Técnicas de Silenciamento de Genes , Fatores de Crescimento de Células Hematopoéticas/genética , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Homeostase/genética , Dados de Sequência Molecular , Ligação Proteica/genética , RNA Interferente Pequeno/genética , Receptores de Laminina/genética , Alinhamento de Sequência , Técnicas do Sistema de Duplo-Híbrido
7.
Ann Hematol ; 92(1): 101-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22971811

RESUMO

Multiple myeloma (MM) is classically illustrated by a desynchronized cytokine system with rise in inflammatory cytokines. There are recent reports which emphasized the potential role of angiogenesis in the development of MM. Role of cyclooxygenase 2 (COX-2) is well documented in the pathogenesis of solid tumors, but little is known about its occurrence and function in hematologic neoplasms. Involvement of neoangiogenesis is reported in the progression of MM, and angiopoietins probably contribute to this progression by enhancing neovascularization. Circulatory and mRNA levels of angiogenic factors and cyclooxygenase were determined in 125 subjects (75 MM patients and 50 healthy controls) by using enzyme-linked immunosorbent assay and quantitative PCR. We observed significant increase for angiogenic factors (Ang-1, Ang-2, hepatocyte growth factor, and vascular endothelial growth factor) and cyclooxygenase at circulatory level, as well as at mRNA level, as compared to healthy controls except insignificant increase for Ang-1 at circulatory level. We have also observed the significant positive correlation of all angiogenic factors with cyclooxygenase. The strong association found between angiogenic factors and COX-2 in this study may lead to the development of combination therapeutic strategy to treat MM. Therefore, targeting COX-2 by using its effective inhibitors demonstrating antiangiogenic and antitumor effects could be used as a new therapeutic approach for treatment of MM.


Assuntos
Angiopoietina-1/biossíntese , Angiopoietina-2/biossíntese , Ciclo-Oxigenase 2/biossíntese , Fatores de Crescimento de Células Hematopoéticas/biossíntese , Mieloma Múltiplo/genética , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/genética , Adulto , Idoso , Inibidores da Angiogênese/uso terapêutico , Angiopoietina-1/sangue , Angiopoietina-1/genética , Angiopoietina-2/sangue , Angiopoietina-2/genética , Ciclo-Oxigenase 2/sangue , Ciclo-Oxigenase 2/genética , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Perfilação da Expressão Gênica , Fatores de Crescimento de Células Hematopoéticas/sangue , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Índia , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/sangue , Mieloma Múltiplo/tratamento farmacológico , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/sangue , RNA Neoplásico/biossíntese , RNA Neoplásico/sangue , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
8.
Int Immunopharmacol ; 13(3): 245-56, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22609473

RESUMO

A stress response can induce myeloid progenitor cell (MPC) proliferation, mobilization, and extramedullary hematopoiesis (EMH) within lymphoid and parenchymal organs. Our studies using in vivo BrdU labeling, Ki-67 IHC staining, and carboxyfluorescein succinimidyl ester (CFSE) adoptive cell transfer revealed that spleens, rather than bone marrow (BM) and peripheral blood (PB), from 4T1 mammary tumor-bearing (TB) mice were the primary site of MPC proliferation. The resultant increase in MPCs was associated with tumor hematopoietic growth factor (GF) transcription, decreased apoptosis, as well as, prolonged survival of splenic MPCs. In naïve mice, i.v. injected CFSE-labeled MDSCs (myeloid-derived suppressor cells) initially accumulated in the lungs, while in TB mice, they rapidly sequestered in the spleen. In contrast, a few of the injected MDSCs and leukocytes arrested, proliferated, or accumulated in the marrow, tumor, or PB of TB mice. However, BrdU labeling revealed a significant demargination of proliferating splenic MPCs into the PB. In tumors, despite high GF transcript levels, we found that a high frequency of MDSCs was apoptotic. In summary, tumor growth and cytokines regulate MPC proliferation, trafficking, accumulation, apoptosis, and survival.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/patologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Transferência Adotiva , Animais , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Citocinas/genética , Feminino , Fatores de Crescimento de Células Hematopoéticas/genética , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Baço/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Transcrição Gênica
9.
J Transl Med ; 9: 158, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21943129

RESUMO

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors to develop in the digestive tract. These tumors are highly resistant to conventional chemotherapy and only the introduction of imatinib mesylate has improved the prognosis of patients. However, Response Evaluation Criteria in Solid Tumors are inappropriate for assessing tumor response, and the histological/pathological response to imatinib is variable, heterogeneous, and does not associate with clinical response. The effects of imatinib on responding GISTs are still being explored, and few studies correlate the clinical response with the histological response after pharmacological treatment. Recently, apoptosis and autophagy were suggested as possible alternative mechanisms of pharmacological response. METHODS: Here, we used a proteomic approach, combined with other analyses, to identify some molecular stromal components related to the response/behavior of resected, high-risk GISTs after neoadiuvant imatinib therapy. RESULTS: Our proteomic results indicate an elevated concentration of Stem Cell Growth Factor (SCGF), a hematopoietic growth factor having a role in the development of erythroid and myeloid progenitors, in imatinib-responsive tumor areas. SCGFα expression was detected by mass spectrometry, immunohistochemistry and/or western blot and attributed to acellular matrix of areas scored negative for KIT (CD117). RT-PCR results indicated that GIST samples did not express SCGF transcripts. The recently reported demonstration by Gundacker et al. 1 of the secretion of SCGF in mature pro-inflammatory dendritic cells would indicate a potential importance of SCGF in tissue inflammatory response. Accordingly, inflammatory infiltrates were detected in imatinib-affected areas and the CD68-positivity of the SCGF-positive and KIT-negative areas suggested previous infiltration of monocytes/macrophages into these regions. Thus, chronic inflammation subsequent to imatinib treatment may determine monocyte/macrophage recruitment in imatinib-damaged areas; these areas also feature prominent tumor-cell loss that is replaced by dense hyalinization and fibrosis. CONCLUSIONS: Our studies highlight a possible role of SCGFα in imatinib-induced changes of GIST structure, consistent with a therapeutic response.


Assuntos
Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/metabolismo , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Lectinas Tipo C/metabolismo , Piperazinas/uso terapêutico , Proteômica/métodos , Pirimidinas/uso terapêutico , Benzamidas , Western Blotting , Linhagem Celular Tumoral , Eletroforese em Gel de Poliacrilamida , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Regulação Neoplásica da Expressão Gênica , Glicosilação , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Lectinas Tipo C/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Células Estromais/metabolismo , Células Estromais/patologia
10.
J Immunol ; 186(4): 2073-9, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21220699

RESUMO

A novel factor, named crustacean hematopoietic factor (CHF), was identified from a library of suppression subtractive hybridization with the aim to find downstream genes of an invertebrate cytokine, astakine 1, in the freshwater crayfish Pacifastacus leniusculus. CHF is a small cysteine-rich protein (∼9 kDa) with high similarity to the N-terminal region of vertebrate CRIM1 in containing an insulin growth factor binding protein variant motif with unknown function. CHF was found to be induced in primary cell cultures of crayfish hematopoietic tissue (Hpt) cells (precursors of crayfish blood cells) after treatment with astakine 1. Silencing of CHF did not affect the renewal of Hpt cells in vitro, but induced apoptosis of Hpt cells. CHF is exclusively expressed in the blood cell lineage of crayfish (Hpt cells and blood cells), and in vivo RNA interference experiments show that knockdown of this gene results in severe loss of blood cells and a higher apoptotic rate in Hpt. Our data further suggest that crayfish CHF is critical for the survival of hemocytes and Hpt cells by preventing their apoptosis, thus it plays an important role in hemocyte homeostasis in crayfish. Our study of CHF may also shed light on the function of this untypical insulin growth factor binding protein motif located in the N-terminal of vertebrate CRIM1.


Assuntos
Astacoidea/citologia , Astacoidea/imunologia , Hematopoese/imunologia , Fatores de Crescimento de Células Hematopoéticas/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/genética , Apoptose/imunologia , Astacoidea/genética , Sequência de Bases , Células Cultivadas , Hibridização Genômica Comparativa , Biblioteca Gênica , Inativação Gênica/imunologia , Hematopoese/genética , Fatores de Crescimento de Células Hematopoéticas/genética , Dados de Sequência Molecular , Técnica de Subtração , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
11.
Infect Immun ; 78(1): 453-60, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19884328

RESUMO

Plasmodium falciparum malaria is a leading global cause of infectious disease burden. In areas in which P. falciparum transmission is holoendemic, such as western Kenya, severe malarial anemia (SMA) results in high rates of pediatric morbidity and mortality. Although the pathophysiological basis of SMA is multifactorial, we recently discovered that suppression of unexplored hematopoietic growth factors that promote erythroid and myeloid colony development, such as stem cell growth factor (SCGF) (C-type lectin domain family member 11A [CLEC11A]), was associated with enhanced development of SMA and reduced erythropoietic responses. To extend these investigations, the relationships between a novel SCGF promoter variant (-539C/T, rs7246355), SMA (hemoglobin [Hb] < 6.0 g/dl), and reduced erythropoietic responses (reticulocyte production index [RPI], <2.0) were investigated with Kenyan children (n = 486) with falciparum malaria from western Kenya. Circulating SCGF was positively correlated with hemoglobin levels (r = 0.251; P = 0.022) and the reticulocyte production index (RPI) (r = 0.268; P = 0.025). Children with SMA also had lower SCGF levels than those in the non-SMA group (P = 0.005). Multivariate logistic regression analyses controlling for covariates demonstrated that individuals with the homologous T allele were protected against SMA (odds ratio, 0.57; 95% confidence interval [95% CI] 0.34 to 0.94; P = 0.027) relative to CC (wild-type) carriers. Carriers of the TT genotype also had higher SCGF levels in circulation (P = 0.018) and in peripheral blood mononuclear cell culture supernatants (P = 0.041), as well as an elevated RPI (P = 0.005) relative to individuals with the CC genotype. The results presented here demonstrate that homozygous T at -539 in the SCGF promoter is associated with elevated SCGF production, enhanced erythropoiesis, and protection against the development of SMA in children with falciparum malaria.


Assuntos
Anemia/etiologia , Anemia/genética , Regulação da Expressão Gênica/fisiologia , Fatores de Crescimento de Células Hematopoéticas/genética , Lectinas Tipo C/genética , Malária Falciparum/complicações , Pré-Escolar , Feminino , Variação Genética , Genótipo , Humanos , Lactente , Masculino , Regiões Promotoras Genéticas
12.
Infect Immun ; 77(9): 3864-71, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19528216

RESUMO

In areas of holoendemic Plasmodium falciparum transmission, severe malarial anemia (SMA) is a leading cause of pediatric morbidity and mortality. Although many soluble mediators regulate erythropoiesis, it is unclear how these factors contribute to development of SMA. Investigation of novel genes dysregulated in response to malarial pigment (hemozoin [PfHz]) revealed that stem cell growth factor (SCGF; also called C-type lectin domain family member 11A [CLEC11A]), a hematopoietic growth factor important for development of erythroid and myeloid progenitors, was one of the most differentially expressed genes. Additional experiments with cultured peripheral blood mononuclear cells (PBMCs) demonstrated that PfHz decreased SCGF/CLEC11A transcriptional expression in a time-dependent manner. Circulating SCGF levels were then determined for Kenyan children (n = 90; aged 3 to 36 months) presenting at a rural hospital with various severities of malarial anemia. SCGF levels in circulation (P = 0.001) and in cultured PBMCs (P = 0.004) were suppressed in children with SMA. Circulating SCGF also correlated positively with hemoglobin levels (r = 0.241; P = 0.022) and the reticulocyte production index (RPI) (r = 0.280; P = 0.029). In addition, SCGF was decreased in children with reduced erythropoiesis (RPI of <2) (P < 0.001) and in children with elevated levels of naturally acquired monocytic PfHz (P = 0.019). Thus, phagocytosis of PfHz promotes a decrease in SCGF gene products, which may contribute to reduced erythropoiesis in children with SMA.


Assuntos
Anemia/etiologia , Fatores de Crescimento de Células Hematopoéticas/genética , Lectinas Tipo C/genética , Malária/complicações , Pré-Escolar , Eritropoese , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Fatores de Crescimento de Células Hematopoéticas/sangue , Fatores de Crescimento de Células Hematopoéticas/fisiologia , Hemeproteínas/fisiologia , Humanos , Lactente , Lectinas Tipo C/sangue , Lectinas Tipo C/fisiologia , Malária/metabolismo , Masculino , Fagocitose , Reticulócitos/fisiologia
13.
Br J Dermatol ; 160(6): 1180-7, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19416273

RESUMO

BACKGROUND: Vitiligo vulgaris is a depigmentary disorder resulting from the disappearance of functional melanocytes. Currently, the pathogenesis of this disorder remains obscure. OBJECTIVES: Genetic analysis of patients with vitilgo may provide important clues for elucidating the complex pathomechanisms involved in the disease process. Because dysfunctional keratinocytes have recently been implicated in the pathogenesis of vitiligo vulgaris, we conducted a case-control association study to investigate this phenomenon. PATIENTS AND METHODS: Fifty-one patients with vitiligo vulgaris and 118 healthy controls from Taiwan were recruited to investigate the association between relevant keratinocyte-related genes and the occurrence of vitiligo vulgaris. This study genotyped 11 single-nucleotide polymorphisms (SNPs) in five genes including stem cell factor (SCF, also known as KITLG), basic fibroblast growth factor (bFGF, also known as NuDT6), endothelin-1 (EDN1), hepatocyte growth factor (HGF) and stem cell growth factor (SCGF, also known as CLEC11A). RESULTS: Our results revealed that the A allele for SNP rs11104947 in the SCF gene and the T allele for SNP rs13866 in the SCGF gene were, respectively, associated with a 1.95- and a 2.14-fold risk of developing vitiligo vulgaris. A higher risk was also detected among subjects who carried the SCF rs995029/rs11104947 C/A haplotype (odds ratio = 2.45). Furthermore, the at-risk alleles for SCF rs11104947 (A allele) and for SCGF SNP rs13866 (T allele) were found to display a 7.92-fold increased gene-gene combined risk. No significant relationship between polymorphic frequency for genes bFGF, EDN1 as well as HGF and occurrence of vitiligo vulgaris was observed. CONCLUSIONS: These novel genetic findings provide new insights in relation to the mechanisms that might be involved in the development of vitiligo vulgaris.


Assuntos
Queratinócitos/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Fator de Células-Tronco/genética , Vitiligo/genética , Adulto , Povo Asiático/genética , Estudos de Casos e Controles , Endotelina-1/genética , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Predisposição Genética para Doença , Fatores de Crescimento de Células Hematopoéticas/genética , Fator de Crescimento de Hepatócito/genética , Humanos , Lectinas Tipo C/genética , Masculino , Pessoa de Meia-Idade , Taiwan , Adulto Jovem
14.
Cytokine Growth Factor Rev ; 20(1): 87-94, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19223217

RESUMO

Hematopoietic growth factor (HGF) mimetics offer a number of attractive advantages as therapeutic agents. Small chemical compounds, in particular, provide reduced cost and oral availability. As many of these mimetics are unrelated in structure to the normal cytokine the immunogenic response is not a significant issue. Isolation of small peptide agonists for erythropoietin (EPO) and thrombopoietin (TPO) receptors has been associated with significant translational challenges and here we summarize approaches used to achieve the potency and stability required for clinical utility. We also compare and contrast the initial screening approaches, and the translational and clinical issues associated with two recently approved TPO mimetics, romiplostim and the orally available eltrombopag. Finally we summarize the development and clinical findings for the EPO mimetic, Hematide, consider alternative approaches, and discuss the future potential for isolation of growth factor (GF) mimetics.


Assuntos
Fatores de Crescimento de Células Hematopoéticas/química , Animais , Proteínas de Transporte/química , Química Farmacêutica/métodos , Citocinas/química , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Peptídeos/química , Polietilenoglicóis/química , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Receptores da Eritropoetina/agonistas , Receptores Fc/química , Receptores de Trombopoetina/agonistas , Proteínas Recombinantes de Fusão , Trombopoetina
15.
Eur J Pharmacol ; 571(2-3): 88-96, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17628529

RESUMO

Histone deacetylase inhibitors (HDAC inhibitors) are an emerging class of anticancer agents. To elucidate the mechanism of HDAC inhibitor-induced thrombocytopenia, we focused on the effects of HDAC inhibitors on megakaryocyte differentiation and performed Affymetrix GeneChip analysis of human megakaryocytic HEL cells treated with or without HDAC inhibitors. Here, we report that GATA-1 and 10 haematopoietic factors (SCL, NF-E2, EKLF, Pleckstrin, Thrombin-R, LMO2, PU.1, Fli-1, AML1, and TCF11) are transcriptionally repressed by HDAC inhibitors in a similar pattern (R>0.98), and putative GATA-1-binding sites are found in almost all promoters of these genes. In addition, luciferase reporter assays reveal that mutations of GATA-1-binding sites in the GATA-1 promoter abolish its sensitivity to HDAC inhibitor-mediated down-regulation in HEL cells. Further, this report also asserts that HDAC inhibitor increases megakaryocyte counts and inhibits GATA-1 gene expression in rat spleen. Together, these results suggest that HDAC inhibitors inhibit GATA-1 gene expression by decreasing the transactivation function of GATA-1 itself, and that this may in turn lead to a delay in megakaryocyte maturation and finally cause thrombocytopenia. Our findings may help our understanding of the molecular mechanism of HDAC inhibitor-mediated GATA-1 transcriptional repression and to reduce the risk of HDAC inhibitor-induced thrombocytopenia.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Fator de Transcrição GATA1/metabolismo , Hematopoese/efeitos dos fármacos , Inibidores de Histona Desacetilases , Megacariócitos/efeitos dos fármacos , Trombocitopenia/metabolismo , Acetilação , Animais , Antineoplásicos/efeitos adversos , Contagem de Células Sanguíneas , Western Blotting , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo , Inibidores Enzimáticos/efeitos adversos , Fator de Transcrição GATA1/genética , Perfilação da Expressão Gênica/métodos , Genes Reporter , Hematopoese/genética , Fatores de Crescimento de Células Hematopoéticas/genética , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Luciferases/genética , Masculino , Megacariócitos/enzimologia , Megacariócitos/metabolismo , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos Cíclicos/farmacologia , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew , Baço/efeitos dos fármacos , Baço/metabolismo , Trombocitopenia/sangue , Trombocitopenia/induzido quimicamente , Trombocitopenia/genética , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Transfecção
16.
Differentiation ; 75(1): 24-34, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17244019

RESUMO

The fate of human hematopoietic stem cells (HSCs)/progenitor cells (HPCs) is influenced by bone marrow (BM) stromal cells. To investigate the role of stromal cells in the hematopoietic support, we have transduced human fetal BM stromal cells (FBMSCs) with a human telomerase catalytic subunit (hTERT). One of the resultant cell lines was identified as osteoblasts, because it contained mineral deposits and constitutively expressed osteogenic genes osteocalcin, osteopontin, collagen type I, osteoblast marker alkaline phosphatase, but not marrow stromal cell marker STRO-1 and CD105. The hTERT-transduced fetal BM-derived osteoblastic cells (FBMOB-hTERT) can actively maintain the capacity of self-renewal and multipotency of HSCs/HPCs at least partly through transcriptional up-regulation of hematopoietic growth factors such as stem cell growth factors (SCFs) and Wnt-5A during interaction with HSCs/HPCs. The enhanced transcription of SCFs and Wnt-5A appears to be mediated by CD29 signaling. Moreover, the FBMOB-hTERT cells seem superior to primary FBMSCs in supporting hematopoiesis, because they are more potent than primary FBMSCs in supporting the ex vivo expansion and long-term culture initiating cells activity of HSCs. The FBMOB-hTERT cell line has been maintained in vitro more than 125 population doublings without tumorigenicity. The results indicate that the FBMOB-hTERT is useful for the study of molecular mechanisms by which osteoblasts support hematopoiesis.


Assuntos
Células da Medula Óssea/citologia , Linhagem Celular/citologia , Hematopoese , Osteoblastos/citologia , Telomerase/genética , Animais , Células da Medula Óssea/enzimologia , Domínio Catalítico , Linhagem Celular/enzimologia , Feto/citologia , Hematopoese/genética , Fatores de Crescimento de Células Hematopoéticas/genética , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Camundongos , Osteoblastos/enzimologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Células Estromais/citologia , Células Estromais/enzimologia , Transdução Genética , Regulação para Cima , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Quinases da Família src
17.
Exp Hematol ; 33(12): 1544-53, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16338498

RESUMO

OBJECTIVE: Gene transfer of the telomerase catalytic subunit (TERT) into primary human stromal cells prolonged their lifespan. However, primary human stromal cells are actually composed of adipocytes, myofibroblasts, osteoblasts, etc. Our objective was to investigate the phenotype and hematopoietic-support of the human telomerized stromal cell (HTS) in clonal level. MATERIALS AND METHODS: We established HTS clones (HTS-1 to HTS-9) from a parental population of HTSs by limiting dilution. Hematopoietic-supporting activity of the HTS clones was examined by coculturing with CD34(+) cells. RESULTS: HTS-1 to HTS-3 contained alkaline phosphatase (ALP)(+) cells, and HTS-4 to HTS-9 were composed of both ALP(+) and alpha-smooth muscle actin-positive cells. Although all HTS clones exhibited normal growth kinetics, one of the HTS clones exhibited a chromosomal abnormality. Moreover, the parental population of the HTS cells acquired an increased growth rate and anchorage independence after 4 years of culturing. Expression of hematopoietic growth factors, such as stem cell factor, angiopoietin-1, and hedgehog mRNA was detected in all HTS clones. The degree of hematopoietic progenitor support differed between the HTS clones, and the expansion level of CD34(+) cells was the highest in HTS-8. CONCLUSION: Human telomerized stromal cell clones exhibited the phenotype of hematopoietic supporting osteoblastic and myofibroblastic cells after long-term culture. Clinical application of HTS cells should be limited because of their potential for neoplastic transformation after hTERT gene transfer. HTS cells may be useful for analyzing the molecular mechanism of hematopoietic support of human stromal cells.


Assuntos
Fibroblastos/citologia , Hematopoese , Osteoblastos/citologia , Células Estromais/citologia , Telômero/genética , Transdução Genética , Células da Medula Óssea , Linhagem da Célula , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Células Clonais , Técnicas de Cocultura , Fatores de Crescimento de Células Hematopoéticas/análise , Fatores de Crescimento de Células Hematopoéticas/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Imunofenotipagem , RNA Mensageiro/análise , Células Estromais/metabolismo , Fatores de Tempo
18.
Biol Neonate ; 88(3): 214-27, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16210844

RESUMO

BACKGROUND: Innate and adaptive immunity is comprised of cellular and humoral factors that provide rapid protection against microbial invasion. However, immaturity of innate and adaptive immune responses in the perinatal period predisposes the neonate to increased infectious morbidity and mortality from a variety of organisms. OBJECTIVES: To elucidate dysregulation of expression of various immunoregulatory and cytokine genes and its association with the immaturity in neonatal phagocytic cellular immunity. METHODS: Comparison of protein production and mRNA of granulocyte macrophage colony-stimulating factor (GM-CSF), macrophage colony-stimulating factor (M-CSF), granulocyte colony-stimulating factor (G-CSF), interleukin (IL)-12, IL-15 and IL-18 in adult peripheral blood (APB) mononuclear cells (MNC) and cord blood (CB) MNC was studied. Effects of hematopoietic growth factors (HGFs, GM-CSF, M-CSF, G-CSF, IL-11) were studied in vivo in rats as well as randomized controlled studies conducted in neonates. Oligonucleotide microarrays were used to study gene expression patterns of activated CB and APB monocytes and dendritic cells. RESULTS AND CONCLUSIONS: We demonstrated dysregulation of various immunoregulatory and cytokine genes in CB MNC. This dysregulation may in part explain the immaturity of neonatal cell-mediated immunity. There are probably various dysregulated cytokines yet to be discovered. Biological agents such as IL-2, IL-12, IL-11 and/or IL-18 alone or in combination with HGFs should be considered for future studies to identify new approaches to enhance neonatal host defense, and thereby decrease the incidence of neonatal sepsis and the consequent high risk of morbidity and mortality.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica , Imunidade Celular/genética , Imunidade/genética , Fagócitos/imunologia , Animais , Animais Recém-Nascidos , Citocinas/fisiologia , Hematopoese , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Recém-Nascido , Células Matadoras Naturais/imunologia , Linfocinas/farmacologia , Fagocitose/genética , Fagocitose/imunologia
19.
Protein Expr Purif ; 43(2): 126-32, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16139754

RESUMO

A novel recombinant dual human stem cell factor (rdhSCF) gene which consisted of a full-length hSCF(1-165aa) cDNA and a truncated hSCF (1-145aa) cDNA, linked by a peptide (GGGGSGGGGSGG) coding region, was constructed and cloned into Escherichia coli expression vector pET-22b. The rdhSCF was expressed at high level in E. coli BL21(DE3) and existed mainly as inclusion bodies. The inclusion bodies were solubilized in urea and refolded by ion-exchange chromatography. After renaturation, the purity of the yielded rdhSCF was up to 90%. Cell proliferation assay showed that the specific activity of the rdhSCF was 2.86x10(5) U/mg, about 1.66 times as high as that of monomer rhSCF expressed in E. coli.


Assuntos
Expressão Gênica , Fatores de Crescimento de Células Hematopoéticas/biossíntese , Fatores de Crescimento de Células Hematopoéticas/química , Dobramento de Proteína , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Escherichia coli/genética , Vetores Genéticos , Fatores de Crescimento de Células Hematopoéticas/genética , Humanos , Corpos de Inclusão/química , Corpos de Inclusão/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes/genética
20.
Cancer Genet Cytogenet ; 162(1): 10-20, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16157195

RESUMO

An association between functional polymorphisms of genes resulting in decreased detoxification of carcinogens or DNA repair and aberrant promoter methylation is an attractive hypothesis in lung carcinogenesis. The genotypes at polymorphic sites of the glutathione S-transferase (GST) M1 (null/wildtype) and P1 (nucleotide 2627 A/G), myeloperoxidase (MPO) (nucleotide -463 G/A), X-ray repair cross-complementing group 1 (XRCC1) (nucleotides 26304 C/T; 28152 G/A), and NADPH quinine oxidoreductase (NQO1) (nucleotide 609 C/T) genes in 75 Chinese patients with non-small cell lung cancer (NSCLC) were characterized with polymerase chain reaction-restriction fragment length polymorphism. Results were correlated with aberrant methylation of the CDKN2A (alias p16(INK4A)), retinoic acid receptor beta (RARB), methylguanine-DNA methyltransferase (MGMT), and death-associated-protein (DAP) kinase genes in the tumors. In comparison with an age-matched control, none of the polymorphisms were associated with increased lung cancer risks. In male patients, however, the MPO -463 GG homozygous state was associated with CDKN2A (alias p16(INK4A)) methylation (odds ratio OR=3.63, 95% confidence interval CI=1.26-10.51), and the XRCC1 26304 T allele in the heterozygous/homozygous state was associated with methylation of CDKN2A (OR=6.13, 95% CI=1.55-24.16) and RARB (OR=7.67, 95% CI=1.62-36.18). In female patients, the GSTP1 G allele in the heterozygous/homozygous state was associated with RARB methylation (OR=18.0, 95% CI=0.76-427.29). These results showed that functional deficiencies in metabolic pathways that protect cells from carcinogen induced DNA damage might be linked to aberrant promoter methylation of the CDKN2A and RARB genes during lung carcinogenesis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas de Ligação a DNA/genética , Genes p16 , Glutationa Transferase/genética , Fatores de Crescimento de Células Hematopoéticas/genética , Isoenzimas/genética , Neoplasias Pulmonares/genética , Polimorfismo Genético , Receptores do Ácido Retinoico/genética , Proteínas Recombinantes de Fusão/genética , Povo Asiático , Metilação de DNA , Feminino , Genótipo , Glutationa S-Transferase pi , Fator Estimulador de Colônias de Granulócitos , Humanos , Interleucina-3 , Masculino , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , Proteínas Recombinantes , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...