Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
Stem Cell Rev Rep ; 18(8): 3021-3032, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35655001

RESUMEN

Female germline stem cells (FGSCs) have been successfully isolated and characterized from postnatal mammalian and human ovarian tissues. However, the effects and mechanisms of action of natural small-molecule compounds on FGSCs are largely unknown. Here, we found that daidzein promoted the viability and proliferation of FGSCs. To elucidate the mechanism underlying this, we performed RNA-Sequence in daidzein-treated FGSCs and controls. The results showed that there were 153 upregulated and 156 downregulated genes in daidzein treatment. We confirmed the expression of some genes related to cell proliferation in the sequencing results by RT-PCR, such as Type C lectin domain family 11 member a (Clec11a), Mucin1 (Muc1), Glutathione peroxidase 3 (Gpx3), and Tet methylcytosine dioxygenase 1 (Tet1). The high expression of Clec11a at the protein level after daidzein treatment was also confirmed by western blotting. Furthermore, recombinant mouse Clec11a (rmClec11a) protein was shown to promote the viability and proliferation of FGSCs. However, knockdown of Clec11a inhibited the viability and proliferation of FGSCs, which could not be rescued by the administration of daidzein. These results indicate that daidzein promoted the viability and proliferation of FGSCs through Clec11a. In addition, both daidzein and rmClec11a activated the Akt signaling pathway in FGSCs. However, Clec11a knockdown inhibited this pathway, which could not be rescued by daidzein administration. Taken together, our findings revealed that daidzein activates the Akt signaling pathway to promote cell viability and proliferation through upregulating Clec11a. This study should deepen our understanding of the developmental mechanism of FGSCs and female infertility.


Asunto(s)
Isoflavonas , Células Madre Oogoniales , Animales , Femenino , Humanos , Ratones , Proliferación Celular , Isoflavonas/farmacología , Isoflavonas/metabolismo , Mamíferos/metabolismo , Oxigenasas de Función Mixta/metabolismo , Oxigenasas de Función Mixta/farmacología , Células Madre Oogoniales/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Regulación hacia Arriba
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34140410

RESUMEN

We previously described a new osteogenic growth factor, osteolectin/Clec11a, which is required for the maintenance of skeletal bone mass during adulthood. Osteolectin binds to Integrin α11 (Itga11), promoting Wnt pathway activation and osteogenic differentiation by leptin receptor+ (LepR+) stromal cells in the bone marrow. Parathyroid hormone (PTH) and sclerostin inhibitor (SOSTi) are bone anabolic agents that are administered to patients with osteoporosis. Here we tested whether osteolectin mediates the effects of PTH or SOSTi on bone formation. We discovered that PTH promoted Osteolectin expression by bone marrow stromal cells within hours of administration and that PTH treatment increased serum osteolectin levels in mice and humans. Osteolectin deficiency in mice attenuated Wnt pathway activation by PTH in bone marrow stromal cells and reduced the osteogenic response to PTH in vitro and in vivo. In contrast, SOSTi did not affect serum osteolectin levels and osteolectin was not required for SOSTi-induced bone formation. Combined administration of osteolectin and PTH, but not osteolectin and SOSTi, additively increased bone volume. PTH thus promotes osteolectin expression and osteolectin mediates part of the effect of PTH on bone formation.


Asunto(s)
Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/farmacología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Hueso Esponjoso/efectos de los fármacos , Hueso Esponjoso/patología , Femenino , Factores de Crecimiento de Célula Hematopoyética/sangre , Factores de Crecimiento de Célula Hematopoyética/deficiencia , Humanos , Lectinas Tipo C/sangre , Lectinas Tipo C/deficiencia , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Osteoporosis/sangre , Premenopausia/sangre , Vía de Señalización Wnt/efectos de los fármacos
3.
Nature ; 591(7850): 438-444, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33627868

RESUMEN

Stromal cells in adult bone marrow that express leptin receptor (LEPR) are a critical source of growth factors, including stem cell factor (SCF), for the maintenance of haematopoietic stem cells and early restricted progenitors1-6. LEPR+ cells are heterogeneous, including skeletal stem cells and osteogenic and adipogenic progenitors7-12, although few markers have been available to distinguish these subsets or to compare their functions. Here we show that expression of an osteogenic growth factor, osteolectin13,14, distinguishes peri-arteriolar LEPR+ cells poised to undergo osteogenesis from peri-sinusoidal LEPR+ cells poised to undergo adipogenesis (but retaining osteogenic potential). Peri-arteriolar LEPR+osteolectin+ cells are rapidly dividing, short-lived osteogenic progenitors that increase in number after fracture and are depleted during ageing. Deletion of Scf from adult osteolectin+ cells did not affect the maintenance of haematopoietic stem cells or most restricted progenitors but depleted common lymphoid progenitors, impairing lymphopoiesis, bacterial clearance, and survival after acute bacterial infection. Peri-arteriolar osteolectin+ cell maintenance required mechanical stimulation. Voluntary running increased, whereas hindlimb unloading decreased, the frequencies of peri-arteriolar osteolectin+ cells and common lymphoid progenitors. Deletion of the mechanosensitive ion channel PIEZO1 from osteolectin+ cells depleted osteolectin+ cells and common lymphoid progenitors. These results show that a peri-arteriolar niche for osteogenesis and lymphopoiesis in bone marrow is maintained by mechanical stimulation and depleted during ageing.


Asunto(s)
Arteriolas , Linfopoyesis , Osteogénesis , Nicho de Células Madre , Tejido Adiposo/citología , Envejecimiento , Animales , Células de la Médula Ósea/citología , Huesos/citología , Femenino , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Linfocitos/citología , Masculino , Ratones , Receptores de Leptina/metabolismo , Factor de Células Madre , Células del Estroma/citología
4.
Cell Rep ; 33(2): 108252, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053358

RESUMEN

Osteogenic suppressors such as Sclerostin not only regulate skeletal development and regeneration but also serve as anti-osteoporosis drug targets. However, very few druggable suppressors have been identified due to limited understanding of the molecular mechanisms governing osteogenesis. Here, we show that fibroblast activation protein (Fap), a serine protease inhibited by the bone growth factor Osteolectin, is an osteogenic suppressor. Genetic deletion of Fap significantly ameliorates limb trabecular bone loss during aging. Pharmacological inhibition of Fap significantly promotes bone formation and inhibits bone resorption in wild-type mice by differentially regulating canonical Wnt and nuclear factor κB (NF-κB) pathways. Pharmacological inhibition of Fap promotes osteoblast differentiation, inhibits osteoclast differentiation, and significantly attenuates osteoporosis in ovariectomized mice. Epistasis analyses in zebrafish show that Osteolectin functions as an endogenous inhibitor of Fap to promote vertebrae mineralization. Taken together, we identify Fap as an important osteogenic suppressor and a potential drug target to treat osteoporosis.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Osteogénesis , Osteoporosis/tratamiento farmacológico , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Resorción Ósea/complicaciones , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Calcificación Fisiológica , Diferenciación Celular , Epistasis Genética , Eliminación de Gen , Células HEK293 , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/complicaciones , Osteoporosis/diagnóstico por imagen , Osteoporosis/patología , Ovariectomía , Péptido Hidrolasas/metabolismo , Unión Proteica , Pez Cebra , Proteínas de Pez Cebra/metabolismo
5.
Theranostics ; 10(5): 2293-2308, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32089743

RESUMEN

Osteoporosis and osteoporotic fractures severely compromise quality of life in elderly people and lead to early death. Human umbilical cord mesenchymal stromal cell (MSC)-derived extracellular vesicles (hucMSC-EVs) possess considerable therapeutic effects in tissue repair and regeneration. Thus, in the present study, we investigated the effects of hucMSC-EVs on primary and secondary osteoporosis and explored the underlying mechanisms. Methods: hucMSCs were isolated and cultured. EVs were obtained from the conditioned medium of hucMSCs and determined by using transmission electron microscopy, dynamic light scattering and Western Blot analyses. The effects of hucMSC-EVs on ovariectomy-induced postmenopausal osteoporosis and tail suspension-induced hindlimb disuse osteoporosis in mouse models were assessed by using microcomputed tomography, biomechanical, histochemical and immunohistochemical, as well as histomorphometric analyses. Proteomic analysis was applied between hucMSC-EVs and hucMSCs to screen the candidate proteins that mediate hucMSC-EVs function. The effects of hucMSC-EVs on osteogenic and adipogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs), and osteoclastogenesis of the macrophage cell line RAW264.7 in vitro were determined by using cytochemical staining and quantitative real-time PCR analysis. Subsequently, the roles of the key protein in hucMSC-EVs-induced regulation on BMSCs and RAW264.7 cells were evaluated. Results: hucMSCs were able to differentiate into osteoblasts, adipocytes or chondrocytes and positively expressed CD29, CD44, CD73 and CD90, but negatively expressed CD34 and CD45. The morphological assessment revealed the typical cup- or sphere-shaped morphology of hucMSC-EVs with diameters predominantly ranging from 60 nm to 150 nm and expressed CD9, CD63, CD81 and TSG101. The systemic administration of hucMSC-EVs prevented bone loss and maintained bone strength in osteoporotic mice by enhancing bone formation, reducing marrow fat accumulation and decreasing bone resorption. Proteomic analysis showed that the potently pro-osteogenic protein, CLEC11A (C-type lectin domain family 11, member A) was very highly enriched in hucMSC-EVs. In addition, hucMSC-EVs enhanced the shift from adipogenic to osteogenic differentiation of BMSCs via delivering CLEC11A in vitro. Moreover, CLEC11A was required for the inhibitory effects of hucMSC-EVs on osteoclast formation. Conclusion: Our results suggest that hucMSC-EVs serve as a critical regulator of bone metabolism by transferring CLEC11A and may represent a potential agent for prevention and treatment of osteoporosis.


Asunto(s)
Huesos/metabolismo , Vesículas Extracelulares/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoporosis/metabolismo , Cordón Umbilical/metabolismo , Adipocitos/metabolismo , Adipogénesis , Animales , Médula Ósea/metabolismo , Diferenciación Celular , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Osteoblastos/metabolismo , Osteogénesis , Osteoporosis/patología , Proteómica , Células RAW 264.7 , Cordón Umbilical/citología , Microtomografía por Rayos X
6.
Nat Commun ; 11(1): 666, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015345

RESUMEN

Inflammatory signals arising from the microenvironment have emerged as critical regulators of hematopoietic stem cell (HSC) function during diverse processes including embryonic development, infectious diseases, and myelosuppressive injuries caused by irradiation and chemotherapy. However, the contributions of cellular subsets within the microenvironment that elicit niche-driven inflammation remain poorly understood. Here, we identify endothelial cells as a crucial component in driving bone marrow (BM) inflammation and HSC dysfunction observed following myelosuppression. We demonstrate that sustained activation of endothelial MAPK causes NF-κB-dependent inflammatory stress response within the BM, leading to significant HSC dysfunction including loss of engraftment ability and a myeloid-biased output. These phenotypes are resolved upon inhibition of endothelial NF-κB signaling. We identify SCGF as a niche-derived factor that suppresses BM inflammation and enhances hematopoietic recovery following myelosuppression. Our findings demonstrate that chronic endothelial inflammation adversely impacts niche activity and HSC function which is reversible upon suppression of inflammation.


Asunto(s)
Células Endoteliales/metabolismo , Hematopoyesis/fisiología , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Animales , Antígenos CD , Médula Ósea , Cadherinas , Femenino , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Inflamación , Masculino , Ratones , Transducción de Señal , Trasplante Autólogo
7.
Handb Exp Pharmacol ; 261: 257-283, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31451971

RESUMEN

This review focuses on certain hematopoietic growth factors that are used as medications in clinical neonatology. It is important to note at the chapter onset that although all of the pharmacological agents mentioned in this review have been approved by the US Food and Drug administration for use in humans, none have been granted a specific FDA indication for neonates. Thus, in a sense, all of the agents mentioned in this chapter could be considered experimental, when used in neonates. However, a great many of the pharmacological agents utilized routinely in neonatology practice do not have a specific FDA indication for this population of patients. Consequently, many of the agents reviewed in this chapter are considered by some practitioners to be nonexperimental and are used when they judge such use to be "best practice" for the disorders under treatment.The medicinal uses of the agents in this chapter vary considerably, between geographic locations, and sometimes even within an institutions. "Consistent approaches" aimed at using these agents in uniform ways in the practice of neonatology are encouraged. Indeed some healthcare systems, and some individual NICUs, have developed written guidelines for using these agents within the practice group. Some such guidelines are provided in this review. It should be noted that these guidelines, or "consistent approaches," must be viewed as dynamic and changing, requiring adjustment and refinement as additional evidence accrues.


Asunto(s)
Factores de Crecimiento de Célula Hematopoyética/química , Neonatología , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Recién Nacido
8.
Exp Cell Res ; 384(1): 111613, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31494095

RESUMEN

The lipotoxicity is considered as one of the risk for diabetes. Here we report C-type lectin domain family 11, member A (Clec11a) as a new regulator in islet playing a protective role in lipotoxicity induced dysfunction. Islet transcriptome sequencing was performed using the high-fat diet induced obesity (DIO) mice model. We found a significant decrease of Clec11a expression in islets of DIO mice compared to normal control mice, which was further confirmed by real-time PCR. Immunostaining demonstrated the localization of the Clec11a protein in mouse islets. Administration of recombinant human Clec11a (rClec11a) protein promoted the proliferation of islet cells and rescued the inhibition of fatty acid on cell proliferation, which involved the activation of Erk signaling pathway. We also found that the rClec11a altered the expression of genes involved in lipid metabolism.


Asunto(s)
Proliferación Celular/fisiología , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Islotes Pancreáticos/metabolismo , Lectinas Tipo C/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Transducción de Señal/fisiología , Transcriptoma/fisiología
9.
PLoS Biol ; 17(7): e3000350, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31265461

RESUMEN

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.


Asunto(s)
Desarrollo Óseo/genética , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen/métodos , Pruebas Genéticas/métodos , Células Madre Embrionarias de Ratones/metabolismo , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Haploidia , Factores de Crecimiento de Célula Hematopoyética/genética , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones , Ratones Noqueados , Relaxina/genética , Relaxina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Science ; 364(6438)2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31023895

RESUMEN

Metabolic health depends on the capacity of adipose tissue progenitor cells to undergo de novo adipogenesis. The cellular hierarchy and mechanisms governing adipocyte progenitor differentiation are incompletely understood. Through single-cell RNA sequence analyses, we show that the lineage hierarchy of adipocyte progenitors consists of distinct mesenchymal cell types that are present in both mouse and human adipose tissues. Cells marked by dipeptidyl peptidase-4 (DPP4)/CD26 expression are highly proliferative, multipotent progenitors. During the development of subcutaneous adipose tissue in mice, these progenitor cells give rise to intercellular adhesion molecule-1 (ICAM1)/CD54-expressing (CD54+) committed preadipocytes and a related adipogenic cell population marked by Clec11a and F3/CD142 expression. Transforming growth factor-ß maintains DPP4+ cell identity and inhibits adipogenic commitment of DPP4+ and CD142+ cells. Notably, DPP4+ progenitors reside in the reticular interstitium, a recently appreciated fluid-filled space within and between tissues, including adipose depots.


Asunto(s)
Adipocitos/citología , Adipogénesis , Tejido Adiposo/citología , Células Madre Mesenquimatosas/citología , Adipocitos/enzimología , Animales , Dipeptidil Peptidasa 4/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/enzimología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Tromboplastina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
11.
Cancer Lett ; 440-441: 47-53, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30312730

RESUMEN

Angiogenesis is regulated by numerous "classic" factors such as vascular endothelial growth factor (VEGF) and many other endogenous "non-classic"peptides, including erythropoietin (Epo), and granulocyte-/granulocyte macrophage colony stimulating factor (G-/GM-CSF). The latter play an important regulatory role in angiogenesis, especially under pathological conditions and constitute a crosslink between angiogenesis and hematopoiesis. This article reviews studies on the ability of hematopoietic cytokines to affect several endothelial cell functions in tumor angiogenesis. These findings in all these studies support the hypothesis formulated at the beginning of this century that a common ancestral cell, the hemangioblast, gives rise to cells of both the endothelial and the hematopoietic lineages.


Asunto(s)
Factores de Crecimiento de Célula Hematopoyética/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Animales , Humanos , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
12.
Biomed Res Int ; 2018: 6435482, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30246025

RESUMEN

INTRODUCTION: Chronic inflammatory response is one of major contributors in the development of hepatocellular carcinoma (HCC). Inflammatory molecules, such as cytokines and growth factors in the circulation, can be useful in the diagnosis and prognosis of the patients. The stem cell growth factor beta (SCGFß), a newly found protein, is a secreted sulfated glycoprotein and it functions as a growth factor for primitive hematopoietic progenitor cells. The level of SCGFß had been reported to be elevated in several cancer types. However, there is very few or even no information on this protein in the study of HCC, even more in clinical studies. METHODS: A multiplex immunoassay panel of 48 cytokines and growth factors were utilized to screen 68 sera from 29 HCC patients at pretreatment (T0), 1 month (T1), and 6 months (T6) after treatment by either radiofrequency ablation (RF) or transarterial chemoembolization (TACE). Treatment response was evaluated according to mRECIST criteria. RESULTS: Immunoassay screening showed that the levels of IL-17, CTACK, TNFα, IL-2Rα, IL-8, and SCGFß were different in Complete Responders (CR) and Nonresponders (NR) groups. At T0 and T1, the SCGFß level was significantly the highest in NR (23.8 and 40.7 ng/mL, respectively), followed by early recurrence (25.4 and 25.0 ng/mL), and CR (6.7 and 5.3 ng/mL), independently from HCV, stages, and treatment type. Low basal SCGFß level was associated with longer disease-free survival compared to high SCGFß. CONCLUSION: In this study, for the first time, we demonstrate that the high level of serum SCGFß at pre- and posttreatment is associated with HCC nonresponsiveness.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Neoplasias Hepáticas/metabolismo , Anciano , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Femenino , Humanos , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Estudios Retrospectivos , Células Madre , Resultado del Tratamiento
13.
Dev Comp Immunol ; 86: 189-195, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29778989

RESUMEN

Serotonin (5-HT) is a conserved monoamine neurotransmitter that has several physiological functions both in vertebrates and invertebrates. In addition to its well-known function in the central nervous system, 5-HT also participates in peripheral system. However, in crustaceans, the knowledge about peripheral functions of 5-HT is limited. In this study, a role of 5-HT in hematopoiesis in crayfish, Pacifastacus leniusculus, was investigated. The presence of 5-HT in crayfish plasma and the effect of 5-HT injection on hemocyte number were examined. The effects of 5-HT on hematopoietic tissue (HPT) cell proliferation and secretion of the hematopoietic cytokine, astakine 1 (Ast 1) were determined in vitro. The results from this study suggest that 5-HT has no direct effect on HPT cell proliferation, but it participates in crayfish hematopoiesis through stimulating Ast 1 cytokine release from crayfish hemocytes, and thereby affects release of new hemocytes into the circulation.


Asunto(s)
Astacoidea/metabolismo , Hematopoyesis/fisiología , Serotonina/metabolismo , Animales , Proliferación Celular/fisiología , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Hemocitos/metabolismo
14.
Semin Immunopathol ; 39(2): 137-152, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27888331

RESUMEN

Dendritic cells (DCs) are specialized immune sentinels that play key role in maintaining immune homeostasis by efficiently regulating the delicate balance between protective immunity and tolerance to self. Although DCs respond to maturation signals present in the surrounding milieu, multiple layers of suppression also co-exist that reduce the infringement of tolerance against self-antigens. These tolerance inducing properties of DCs are governed by their origin and a range of other factors including distribution, cytokines, growth factors, and transcriptional programing, that collectively impart suppressive functions to these cells. DCs directing tolerance secrete anti-inflammatory cytokines and induce naïve T cells or B cells to differentiate into regulatory T cells (Tregs) or B cells. In this review, we provide a detailed outlook on the molecular mechanisms that induce functional specialization to govern central or peripheral tolerance. The tolerance-inducing nature of DCs can be exploited to overcome autoimmunity and rejection in graft transplantation.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Tolerancia Inmunológica , Animales , Presentación de Antígeno/inmunología , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/metabolismo , Diferenciación Celular , Reactividad Cruzada/inmunología , Citocinas/metabolismo , Células Dendríticas/clasificación , Células Dendríticas/citología , Regulación de la Expresión Génica , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos , Inmunomodulación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Especificidad de Órganos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Vacunas/inmunología
15.
Elife ; 52016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27976999

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/fisiología , Osteogénesis , Esqueleto/fisiología , Adulto , Animales , Células Cultivadas , Factores de Crecimiento de Célula Hematopoyética/deficiencia , Factores de Crecimiento de Célula Hematopoyética/genética , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
PLoS One ; 11(12): e0167437, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28005985

RESUMEN

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.


Asunto(s)
Adalimumab/uso terapéutico , Antiinflamatorios/uso terapéutico , Psoriasis/tratamiento farmacológico , Piel/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Adalimumab/farmacología , Adulto , Anciano , Antiinflamatorios/farmacología , Citocinas/genética , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Factores de Crecimiento de Célula Hematopoyética/genética , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Análisis de Componente Principal , Psoriasis/genética , Psoriasis/patología , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Piel/patología , Factores de Tiempo
17.
Exp Hematol ; 44(7): 635-40, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27090409

RESUMEN

The critical human cells that produce neutrophils and platelets within 3 weeks in recipients of hematopoietic transplants are thought to produce these mature blood cells with the same kinetics in sublethally irradiated immunodeficient mice. Quantification of their numbers indicates their relative underrepresentation in cord blood (CB), likely explaining the clinical inadequacy of single CB units in rescuing hematopoiesis in myelosuppressed adult patients. We here describe that exposure of CD34(+) CB cells ex vivo to growth factors that markedly expand their numbers and colony-forming cell content also rapidly (within 24 hours) produce a significant and sustained net loss of their original short-term repopulating activity. This loss of short-term in vivo repopulating activity affects early platelet production faster than early neutrophil output, consistent with their origin from distinct input populations. Moreover, this growth factor-mediated loss is not abrogated by published strategies to increase progenitor homing despite evidence that the effect on rapid neutrophil production is paralleled in time and amount by a loss of the homing of their committed clonogenic precursors to the bone marrow. These results highlight the inability of in vitro or phenotype assessments to reliably predict clinical engraftment kinetics of cultured CB cells.


Asunto(s)
Plaquetas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mielopoyesis , Neutrófilos/metabolismo , Trombopoyesis , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Supervivencia de Injerto , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Factores de Crecimiento de Célula Hematopoyética/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Mielopoyesis/efectos de los fármacos , Trombopoyesis/efectos de los fármacos
18.
Cell Transplant ; 25(5): 863-82, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26787093

RESUMEN

Neural stem cells (NSCs) persist in the subventricular zone lining the ventricles of the adult brain. The resident stem/progenitor cells can be stimulated in vivo by neurotrophic factors, hematopoietic growth factors, magnetic stimulation, and/or physical exercise. In both animals and humans, the differentiation and survival of neurons arising from the subventricular zone may also be regulated by the trophic factors. Since stem/progenitor cells present in the adult brain and the production of new neurons occurs at specific sites, there is a possibility for the treatment of incurable neurological diseases. It might be feasible to induce neurogenesis, which would be particularly efficacious in the treatment of striatal neurodegenerative conditions such as Huntington's disease, as well as cerebrovascular diseases such as ischemic stroke and cerebral palsy, conditions that are widely seen in the clinics. Understanding of the molecular control of endogenous NSC activation and progenitor cell mobilization will likely provide many new opportunities as therapeutic strategies. In this review, we focus on endogenous stem/progenitor cell activation that occurs in response to exogenous factors including neurotrophic factors, hematopoietic growth factors, magnetic stimulation, and an enriched environment. Taken together, these findings suggest the possibility that functional brain repair through induced neurorestoration from endogenous stem cells may soon be a clinical reality.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Ventrículos Cerebrales/citología , Trastornos Cerebrovasculares/terapia , Regeneración Nerviosa/fisiología , Células-Madre Neurales/citología , Enfermedades Neurodegenerativas/terapia , Neurogénesis/fisiología , Ejercicio Físico , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Factores de Crecimiento Nervioso/metabolismo , Estimulación Magnética Transcraneal
19.
Adv Exp Med Biol ; 843: 173-213, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25956299

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Blastocisto/metabolismo , Desarrollo Embrionario/genética , Trompas Uterinas/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Útero/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Blastocisto/citología , Supervivencia Celular , Femenino , Fertilización , Regulación del Desarrollo de la Expresión Génica , Factores de Crecimiento de Célula Hematopoyética/genética , Humanos , Embarazo , Transducción de Señal
20.
Dev Comp Immunol ; 51(1): 39-47, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25720979

RESUMEN

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.


Asunto(s)
Hemocitos/fisiología , Penaeidae/inmunología , Receptores de Laminina/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Técnicas de Silenciamiento del Gen , Factores de Crecimiento de Célula Hematopoyética/genética , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Homeostasis/genética , Datos de Secuencia Molecular , Unión Proteica/genética , ARN Interferente Pequeño/genética , Receptores de Laminina/genética , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...