Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Int J Mol Sci ; 22(12)2021 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-34198626

RESUMEN

Human stem-cell factor (hSCF) stimulates the survival, proliferation, and differentiation of hematopoietic cells by binding to the c-Kit receptor. Various applications of hSCF require the efficient and reliable production of hSCF. hSCF exists in three forms: as two membrane-spanning proteins hSCF248 and hSCF229 and truncated soluble N-terminal protein hSCF164. hSCF164 is known to be insoluble when expressed in Escherichia coli cytoplasm, requiring a complex refolding procedure. The activity of hSCF248 has never been studied. Here, we investigated novel production methods for recombinant hSCF164 and hSCF248 without the refolding process. To increase the solubility of hSCF164, maltose-binding protein (MBP) and protein disulfide isomerase b'a' domain (PDIb'a') tags were attached to the N-terminus of hSCF164. These fusion proteins were overexpressed in soluble form in the Origami 2(DE3) E. coli strain. These solubilization effects were enhanced at a low temperature. His-hSCF248, the poly-His tagged form of hSCF248, was expressed in a highly soluble form without a solubilization tag protein, which was unexpected because His-hSCF248 contains a transmembrane domain. hSCF164 was purified using affinity and ion-exchange chromatography, and His-hSCF248 was purified by ion-exchange and gel filtration chromatography. The purified proteins stimulated the proliferation of TF-1 cells. Interestingly, the EC50 value of His-hSCF248 was 1 pg/mL, 100-fold lower than 9 ng/mL hSCF164. Additionally, His-hSCF248 decreased the doubling time, increased the proportion of S and G2/M stages in the cell cycle, and increased the c-Myc expression at a 1000-fold lower concentration than hSCF164. In conclusion, His-hSCF248 was expressed in a soluble form in E. coli and had stronger activity than hSCF164. The molecular chaperone, MBP, enabled the soluble overexpression of hSCF164.


Asunto(s)
Factor de Células Madre/biosíntesis , Secuencia de Aminoácidos , Ciclo Celular , Proliferación Celular , Regulación de la Expresión Génica , Humanos , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad , Factor de Células Madre/química
2.
Pathol Oncol Res ; 25(2): 611-624, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30402808

RESUMEN

The initiation of prostatic malignancy has been linked to chronic inflammation. Stem cell factor (SCF) is an inflammatory cytokine that is specific to the c-KIT receptor which is type III receptor tyrosine kinase (RTK). Cyclooxygenases (COXs) are the main enzymes which are responsible for prostaglandins production from arachidonic acid. COX2 is an enzyme which is produced under different pathological conditions. The aim of our study; is to investigate the clinicopathological and the prognostic significance of SCF and COX-2 expression in prostatic adenocarcinoma (PC), chronic prostatitis and nodular prostatic hyperplasia (NPH) in a trial to clarify the role of inflammation as a risk factor for prostatic carcinogenesis and cancer progression. SCF and COX-2 tissue protein expression were evaluated in 50 cases of PC, 20 cases of chronic prostatitis and 10 cases of NPH using immunohistochemistry, patients were followed up for 5 years. The relationship between their levels of expressions, clinicopathological, and prognostic criteria were studied. SCF expression in PC was positively correlated with advanced patient age (p = <0.001), high level of PSA (p = 0.010), higher Gleason score (p = 0.011). COX-2 expression in PC was positively correlated with advanced patient age (p = <0.001), high level of PSA (p = 0.016), advanced D'Amico risk group (p = 0.038). High levels of expression of both SCF& COX-2 are associated with higher incidence of tumor relapse, worse disease overall survival and free survival (p < 0.001). SCF and COX-2 are associated with PC progression and associated with poor prognosis in PC patients.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/análisis , Ciclooxigenasa 2/biosíntesis , Neoplasias de la Próstata/patología , Factor de Células Madre/biosíntesis , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adulto , Anciano , Carcinogénesis/metabolismo , Supervivencia sin Enfermedad , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/mortalidad , Prostatitis/metabolismo , Prostatitis/patología
3.
Medicina (Kaunas) ; 54(5)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463207

RESUMEN

Background and objectives: Paocai (pickled cabbage), which is fermented by lactic acid bacteria, is a traditional Chinese food. The microorganisms of Paocai were isolated and identified, and the constipation inhibition effect of one of the isolated Lactobacillus was investigated. Materials and Methods: The 16S rDNA technology was used for microbial identification. A mouse constipation model was established using activated carbon. After intragastric administration of Lactobacillus (108 CFU/mL), the mice were dissected to prepare pathological sections of the small intestine. Serum indicators were detected using kits, and the expression of small intestine-related mRNAs was detected by qPCR assay. Results: One strain of Lactobacillus was identified and named Lactobacillus fermentum CQPC03 (LF-CQPC03). Body weight and activated carbon propulsion rate were all higher in mice intragastrically administered with LF-CQPC03 compared with the control group, while the time to the first black stool in treated mice was lower than that in the control group. Serum assays showed that gastrin (Gas), endothelin (ET), and acetylcholinesterase (AchE) levels were significantly higher in the LF-CQPC03-treated mice than in the control group, while somatostatin (SS) levels were significantly lower than in the control mice. Mouse small intestine tissue showed that c-Kit, stem cell factor (SCF), and glial cell-derived neurotrophic factor (GDNF) mRNA expression levels were significantly higher in the LF-CQPC03 treated mice than in control mice, while transient receptor potential cation channel subfamily V member 1 (TRPV1) and inducible nitric oxide synthase (iNOS) expression levels were significantly lower in the LF-CQPC03 treated mice than in control mice. Conclusions: There is a better effect with high-dose LF-CQPC03, compared to the lower dose (LF-CQPC03-L), showing good probiotic potential, as well as development and application value.


Asunto(s)
Brassica/microbiología , Estreñimiento/prevención & control , Alimentos Fermentados/microbiología , Limosilactobacillus fermentum/aislamiento & purificación , Limosilactobacillus fermentum/metabolismo , Probióticos/administración & dosificación , Acetilcolinesterasa/sangre , Animales , Peso Corporal , Carbono/farmacología , Estreñimiento/sangre , Estreñimiento/inducido químicamente , Defecación , Modelos Animales de Enfermedad , Endotelinas/sangre , Heces , Femenino , Fermentación , Gastrinas/sangre , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Intestino Delgado/metabolismo , Intestino Delgado/patología , Ratones , Ratones Endogámicos ICR , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Probióticos/aislamiento & purificación , Probióticos/metabolismo , Somatostatina/sangre , Factor de Células Madre/biosíntesis , Canales Catiónicos TRPV/biosíntesis
4.
Exp Hematol ; 64: 59-70.e2, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29775645

RESUMEN

Mesenchymal stromal cells (MSCs) support hematopoietic stem cells (HSCs) in vivo and enhance HSC engraftment and hematopoietic recovery upon cotransplantation with HSCs. These data have led to the hypothesis that MSCs may affect the HSC niche, leading to changes in HSC retention and trafficking. We studied the effect of MSC administration on the HSC compartment in the bone marrow (BM) in mice. After injection of MSCs, HSC numbers in the BM were decreased coinciding with an increased cell cycle activity compared with phosphate-buffered saline (PBS)-injected controls. Furthermore, the frequency of macrophages was significantly reduced and niche factors including Cxcl12, Scf, and Vcam were downregulated in endosteal cells. These BM changes are reminiscent of events associated with granulocyte colony-stimulating factor (G-CSF)-induced hematopoietic stem and progenitor cell (HSPC) mobilization. Interestingly, coadministration of MSCs and G-CSF resulted in a twofold increase in peripheral blood HSPC release compared with injection of G-CSF alone, whereas injection of MSCs alone did not induce HSPC mobilization. After intravenous administration, MSCs were only observed in the lungs, suggesting that they exert their effect on the HSC niche through a soluble mediator. Therefore, we tested the hypothesis that MSC-derived extracellular vesicles (EVs) are responsible for the observed changes in the HSC niche. Indeed, administration of EVs resulted in downregulation of Cxcl12, Scf, and Vcam and enhanced G-CSF-induced HSPC mobilization at similar levels as MSCs and G-CSF. Together, these data indicate that MSCs induce a permissive state in the BM, enhancing HSPC mobilization through the release of EVs.


Asunto(s)
Médula Ósea/fisiología , Vesículas Extracelulares/fisiología , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Animales , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Citocinas/farmacología , Regulación de la Expresión Génica , Humanos , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología , Factor de Células Madre/biosíntesis , Factor de Células Madre/genética , Nicho de Células Madre , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética
5.
Angiogenesis ; 21(2): 229-236, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29330760

RESUMEN

Receptor tyrosine kinase c-Kit and its ligand stem cell factor (SCF) regulate resident vascular wall cells and recruit circulating progenitors. We tested whether SCF may be induced by oxidized palmitoyl-arachidonoyl-phosphatidylcholine (OxPAPC) known to accumulate in atherosclerotic vessels. Gene expression analysis demonstrated OxPAPC-induced upregulation of SCF mRNA and protein in different types of endothelial cells (ECs). Elevated levels of SCF mRNA were observed in aortas of ApoE-/- knockout mice. ECs produced biologically active SCF because conditioned medium from OxPAPC-treated cells stimulated activation (phosphorylation) of c-Kit in naïve ECs. Induction of SCF by OxPAPC was inhibited by knocking down transcription factor NRF2. Inhibition or stimulation of NRF2 by pharmacological or molecular tools induced corresponding changes in SCF expression. Finally, we observed decreased levels of SCF mRNA in aortas of NRF2 knockout mice. We characterize OxPLs as a novel pathology-associated stimulus inducing expression of SCF in endothelial cells. Furthermore, our data point to transcription factor NRF2 as a major mediator of OxPL-induced upregulation of SCF. This mechanism may represent one of the facets of pleiotropic action of NRF2 in vascular wall.


Asunto(s)
Aorta/metabolismo , Regulación de la Expresión Génica , Factor 2 Relacionado con NF-E2/metabolismo , Fosfatidilcolinas/metabolismo , Factor de Células Madre/biosíntesis , Animales , Aorta/patología , Apolipoproteínas E/deficiencia , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Ratones , Ratones Noqueados para ApoE , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Fosfatidilcolinas/genética , Factor de Células Madre/genética
6.
Cell Tissue Res ; 370(3): 427-440, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28986650

RESUMEN

Homeostatic cell turnover has been extensively characterized in mammals. In their adult tissues, lost or aging differentiated cells are replenished by a self-renewing cohort of stem cells. The stem cells have been particularly well studied in the intestine and are clearly identified by the expression of marker genes including Lgr5 and Bmi1. It is, however, unknown if the established principles of tissue renewal learned from mammals would be operating in non-mammalian systems. Here, we study homeostatic cell turnover in the sea cucumber digestive tube, the organ with high tissue plasticity even in adult animals. Both the luminal epithelium and mesothelium express orthologs of mammalian Lgr5 and Bmi1. However, unlike in mammals, there is no segregation of these positively labeled cells to specific regions in the luminal epithelium, where most of the cell proliferation would take place. In the mesothelium, the cells expressing the stem cell markers are tentatively identified as peritoneocytes. There are significant differences among the five anatomical gut regions in cell renewal dynamics and stem factor expression. The cloaca differs from the rest of the digestive tube as the region with the highest expression of the Lgr5 ortholog, lowest level of Bmi1 and the longest retention of BrdU-labeled cells.


Asunto(s)
Células Epiteliales/metabolismo , Tracto Gastrointestinal/metabolismo , Complejo Represivo Polycomb 1/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Pepinos de Mar/metabolismo , Factor de Células Madre/biosíntesis , Células Madre/metabolismo , Animales , Proliferación Celular , Epitelio/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/genética , Complejo Represivo Polycomb 1/genética , Receptores Acoplados a Proteínas G/genética , Células Madre/citología
7.
Biol Pharm Bull ; 40(9): 1506-1514, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28867733

RESUMEN

Gastric motility disturbance is commonly found in long-standing hyperglycemia. Both delayed and rapid gastric emptying has been reported in diabetes. However, very few studies have followed the changes in gastric emptying during disease progression in diabetes because of technical limitations. 13C-Acetic acid breath test is a validated method which is non-invasive and can be used repeatedly or serially to evaluate gastric emptying changes in animal. We investigated the gastric emptying changes in different stages of diabetes using 13C-acetic acid breath test, as well as its related mechanisms involving interstitial cells of Cajal (ICCs), and stem cell factor (SCF) in streptozotocin-induced diabetic rats. The results showed that gastric emptying was accelerated at the early stage (12 weeks of diabetes) whereas intramuscular ICCs (ICC-IM) networks were not different from normal group. At long-term stage (28 weeks of diabetes), gastric emptying had returned to normal pattern with no delayed. ICC-IM networks were decreased in the diabetic group compared to 12th weeks, and were lower than in the normal group at the same time point. SCF levels were constantly high in the diabetic group than in the normal group. This result indicated that 13C-acetic acid breath test is useful to track the alteration in gastric emptying during disease progression. The change of gastric emptying was not found to be significantly associated with ICC-IM. Elevated SCF may help to preserve ICC-IM, especially in the early phase of diabetes.


Asunto(s)
Ácido Acético/análisis , Pruebas Respiratorias/métodos , Diabetes Mellitus Experimental/fisiopatología , Vaciamiento Gástrico , Animales , Glucemia/análisis , Peso Corporal , Recuento de Células , Diabetes Mellitus Experimental/patología , Progresión de la Enfermedad , Células Intersticiales de Cajal/patología , Masculino , Monitoreo Fisiológico , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Ratas , Ratas Sprague-Dawley , Factor de Células Madre/biosíntesis
8.
Genet Mol Res ; 16(1)2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28290616

RESUMEN

Stem cell factor (SCF), an important stem cell cytokine, has multiple functions. Fibroblasts (FBs), mature mast cells, endothelial cells (ECs), and eosinophil granulocytes can produce SCF in the inflammatory process. Therefore, we aimed to observe SCF expression in FBs, ECs, and macrophages (MPs) in periapical tissues in human chronic periapical disease and investigate the effects of cells expressing SCF in pathogenesis of the disease. Healthy (N = 20), periapical cyst (N = 15), and periapical granuloma (N = 15) tissues were fixed in 10% formalin for 48 h, embedded in paraffin, and stained with hematoxylin and eosin to observe histological changes. SCF expression was observed in FBs, ECs, and MPs in periapical tissues by double immunofluorescence. CD334, CD31, and CD14 are specific markers of FBs, ECs, and MPs, respectively. Results showed that densities of CD334-SCF double-positive FBs, CD31-SCF double-positive ECs, and CD14-SCF double-positive MPs were significantly increased in periapical tissue groups (P < 0.01). There were no significant differences in CD334-SCF double-positive FB and CD31-SCF double-positive EC levels between the two periapical tissue groups (P > 0.05). CD14-SCF double-positive MP density was considerably higher in periapical granulomas than in cysts (P < 0.01). FB, EC, and MP levels were significantly high and densities of CD334-SCF double-positive FBs, CD31-SCF double-positive ECs, and CD14-SCF double-positive MPs improved considerably in chronic periapical tissues, suggesting that the cells might be related to occurrence, development, and pathogenesis of chronic periapical disease.


Asunto(s)
Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Macrófagos/metabolismo , Enfermedades Periapicales/metabolismo , Tejido Periapical/metabolismo , Factor de Células Madre/biosíntesis , Adulto , Anciano , Citocinas/metabolismo , Células Endoteliales/patología , Femenino , Fibroblastos/patología , Humanos , Macrófagos/patología , Masculino , Mastocitos/metabolismo , Mastocitos/patología , Persona de Mediana Edad , Enfermedades Periapicales/patología , Granuloma Periapical/metabolismo , Granuloma Periapical/patología , Tejido Periapical/patología , Quiste Radicular/metabolismo , Quiste Radicular/patología , Factor de Células Madre/metabolismo
9.
Cancer Epidemiol Biomarkers Prev ; 26(2): 197-205, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27742669

RESUMEN

BACKGROUND: The transcription factor high-mobility AT-hook 2 (HMGA2) is involved in stem cell renewal and is expressed in many tumor tissues. Head and neck squamous cell carcinomas (HNSCC) comprise tumors of the upper aerodigestive tract and are characterized by high recurrence rates that represent a challenge to patient management. The study addresses the potential of HMGA2 as a molecular biomarker for HNSCC patient survival. METHODS: Patients with HNSCC of the larynx, pharynx, tonsils, or oral cavity were recruited in a hospital-based case-control study (n = 202). Quantitative expression of HMGA2 in tumor tissues was measured by RT-PCR. In a 6- to 10-year follow-up, secondary cancers, vital status, and cause of death were ascertained. The HR and 95% confidence intervals (CI) for overall, tumor-specific, and progression-free survival were estimated by Cox proportional hazards with HMGA2 expression level as the independent variable. RESULTS: High HMGA2 expression in tumor tissues of HNSCC patients was significantly correlated with negative HPV status (P = 0.01), and associated with shorter overall survival time. In Cox regression modeling, HMGA2 expression yielded a risk increase for overall and tumor-specific death in subsets of HNSCC patients, that is, laryngeal cancer patients (overall survival: HR = 4.00; 95% CI, 1.18-13.62) and in oral cancer patients (tumor-specific survival: HR = 2.88; 95% CI, 1.06-7.84), but not in patients with pharyngeal and tonsillar HNSCC. CONCLUSIONS: HMGA2 expression is associated with a risk increase for adverse outcomes in patients with HNSCC of the larynx and oral cavity. IMPACT: The understanding of stem cell signaling in HNSCC may offer new strategies for cancer treatment. Cancer Epidemiol Biomarkers Prev; 26(2); 197-205. ©2016 AACR.


Asunto(s)
Carcinoma de Células Escamosas/genética , Predicción , Proteína HMGA2/genética , Neoplasias de Cabeza y Cuello/genética , ARN Neoplásico/genética , Factor de Células Madre/genética , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/metabolismo , ADN Viral/análisis , Femenino , Estudios de Seguimiento , Alemania/epidemiología , Proteína HMGA2/biosíntesis , Neoplasias de Cabeza y Cuello/epidemiología , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Morbilidad/tendencias , Papillomaviridae/genética , Reacción en Cadena de la Polimerasa , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello , Factor de Células Madre/biosíntesis , Tasa de Supervivencia/tendencias
10.
J Allergy Clin Immunol ; 139(4): 1205-1216.e6, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27746235

RESUMEN

BACKGROUND: Mast cell (MC) progenitors leave the bone marrow, enter the circulation, and settle in the skin and other tissues. Their maturation in tissues is influenced by the surrounding microenvironment. OBJECTIVE: We tested the hypothesis that environmental factors play a role in MC maturation in the skin. METHODS: MCs were numerically, phenotypically, and functionally compared between germ-free (GF), specific pathogen-free, and GF mice reconstituted with microbiota. The maturity of MCs was then correlated with skin levels of stem cell factor (SCF), a critical MC differentiation factor, and lipoteichoic acid (LTA), a Toll-like receptor 2 ligand. MCs were also evaluated in mice with keratinocyte-specific deletion of Scf. RESULTS: We found that GF mice express abnormally low amounts of SCF, a critical MC differentiation factor, and contain MCs that are largely undifferentiated. Reconstituting the GF microbiota reverted this MC phenotype to normal, indicating that the phenotype is related to ongoing interactions of the microbiota and skin. Consistent with the immaturity of GF MCs, degranulation-provoking compound 48/80 induced less edema in the skin of GF mice than in conventional mice. Our results show that the skin microbiome drives SCF production in keratinocytes, which triggers the differentiation of dermal MCs. Because the skin microbiome is a rich source of LTA, a Toll-like receptor 2 ligand, we mimicked the GF microbiome's effect on MCs by applying LTA to the skin of GF mice. We also demonstrated that MC migration within the skin depends exclusively on keratinocyte-produced SCF. CONCLUSION: This study has revealed a novel mechanism by which the skin microbiota signals the recruitment and maturation of MCs within the dermis through SCF production by LTA-stimulated keratinocytes.


Asunto(s)
Diferenciación Celular/fisiología , Queratinocitos/metabolismo , Mastocitos/citología , Piel/microbiología , Factor de Células Madre/biosíntesis , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Vida Libre de Gérmenes , Humanos , Captura por Microdisección con Láser , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Reacción en Cadena en Tiempo Real de la Polimerasa , Piel/citología , Piel/metabolismo , Ácidos Teicoicos/farmacología
11.
Circ Res ; 119(8): 921-30, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27481956

RESUMEN

RATIONALE: Culture-expanded cells originating from cardiac tissue that express the cell surface receptor cKit are undergoing clinical testing as a cell source for heart failure and congenital heart disease. Although accumulating data support that mesenchymal stem cells (MSCs) enhance the efficacy of cardiac cKit(+) cells (CSCs), the underlying mechanism for this synergistic effect remains incompletely understood. OBJECTIVE: To test the hypothesis that MSCs stimulate endogenous CSCs to proliferate, migrate, and differentiate via the SDF1/CXCR4 and stem cell factor/cKit pathways. METHODS AND RESULTS: Using genetic lineage-tracing approaches, we show that in the postnatal murine heart, cKit(+) cells proliferate, migrate, and form cardiomyocytes, but not endothelial cells. CSCs exhibit marked chemotactic and proliferative responses when cocultured with MSCs but not with cardiac stromal cells. Antagonism of the CXCR4 pathway with AMD3100 (an SDF1/CXCR4 antagonist) inhibited MSC-induced CSC chemotaxis but stimulated CSC cardiomyogenesis (P<0.0001). Furthermore, MSCs enhanced CSC proliferation via the stem cell factor/cKit and SDF1/CXCR4 pathways (P<0.0001). CONCLUSIONS: Together these findings show that MSCs exhibit profound, yet differential, effects on CSC migration, proliferation, and differentiation and suggest a mechanism underlying the improved cardiac regeneration associated with combination therapy using CSCs and MSCs. These findings have important therapeutic implications for cell-based therapy strategies that use mixtures of CSCs and MSCs.


Asunto(s)
Quimiocina CXCL12/biosíntesis , Células Madre Mesenquimatosas/fisiología , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Receptores CXCR4/biosíntesis , Factor de Células Madre/biosíntesis , Animales , Animales Recién Nacidos , Movimiento Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Ratones , Ratones Transgénicos , Proyectos Piloto , Transducción de Señal/fisiología , Porcinos
12.
Invest Ophthalmol Vis Sci ; 57(3): 1201-6, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26978025

RESUMEN

PURPOSE: Stem cell factor (SCF) has recently demonstrated activity as a novel endothelial permeability factor that contributes to the development of diabetes-induced hyperpermeable retinal vasculature. This study investigated the therapeutic potential of masitinib, a pharmacologic inhibitor of the SCF receptor cKit, for prevention of diabetes-induced breakdown of blood retinal barrier (BRB). METHODS: Permeability assays were performed with human retinal microvascular endothelial cells (HRMECs) and murine retinal vasculature. Localization of vascular endothelial (VE)-cadherin and activation of SCF signaling pathway was determined by immunofluorescence and Western blotting assays. Mice and rats with streptozotocin (STZ)-induced diabetes were used to investigate the role of cKit and masitinib in diabetes-induced retinal vascular hyperpermeability. RESULTS: Masitinib substantially blocked SCF-induced phosphorylation of cKit in HRMECs. In vitro and in vivo vascular permeability assays showed that masitinib significantly inhibited SCF-induced endothelial hyperpermeability and junctional loss of VE-cadherin. Streptozotocin-induced diabetes was induced in cKit-mutant mice with low cKit expression in their endothelial cells. Although diabetic wild-type mice exhibited enhanced retinal vascular leakage, diabetic cKit-mutant mice showed no increase in retinal vascular leakage or alteration in the distribution of VE-cadherin; this indicates the crucial role of cKit in diabetes-induced breakdown of BRB. Moreover, in vivo prevention experiments showed that an intravitreal injection of masitinib substantially inhibited the development of hyperpermeable retinal vasculature. CONCLUSIONS: These results provide the first demonstration that cKit inhibitors, such as masitinib, might be promising therapeutics for prevention of diabetes-induced breakdown of the BRB.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Diabetes Mellitus Experimental , Retinopatía Diabética/complicaciones , Endotelio Vascular/efectos de los fármacos , Enfermedades de la Retina/prevención & control , Tiazoles/administración & dosificación , Resistencia Vascular/efectos de los fármacos , Animales , Benzamidas , Barrera Hematorretinal/efectos de los fármacos , Western Blotting , Células Cultivadas , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Piperidinas , Inhibidores de Proteínas Quinasas/administración & dosificación , Piridinas , ARN/genética , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/etiología , Enfermedades de la Retina/metabolismo , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/biosíntesis , Factor de Células Madre/genética
13.
PLoS One ; 11(2): e0149031, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26862759

RESUMEN

It is well known that natriuretic peptides (NPs) are involved in the regulation of gastrointestinal motility. Interstitial cells of Cajal (ICC) are the pacemaker cells of gastrointestinal motility and gastrointestinal dyskinesia is one of the important digestive tract symptoms of depression. However, it is unclear whether they are involved in depression-induced loss of ICC. The aim of the present study was to investigate the relationship between the natriuretic peptide signaling pathway and depression-induced loss of gastric ICC in depressed rats. These results showed that the expression of c-kit and stem cell factor (SCF) in smooth muscle layers of stomach were down-regulated in depressed rats at the mRNA and protein levels. The expression of natriuretic peptide receptor (NPR)-A, B and C were up-regulated in the stomach of depressed rats at the mRNA and protein levels. NPR-A, B and C can significantly decrease the expression of SCF to treat cultured gastric smooth muscle cells (GSMCs) obtained from normal rats with different concentrations of C-type natriuretic peptide (CNP). Pretreatment of cultured GSMCs with 8-Brom-cGMP (8-Br-cGMP, a membrane permeable cGMP analog), cANF (a specific NPR-C agonist) and CNP (10-6 mol/L) demonstrated that 8-Br-cGMP had a similar effect as CNP, but treatment with cANF did not. The results of the methyl thiazolyl tetrazolium bromide (MTT) assay indicated that high concentrations of cANF (10-6 mol/L) restrained the proliferation of cultured GSMCs. Taken together, these results indicate that the up-regulation of the NPs/NPR-C and NPs/NPR-A, B/cGMP signaling pathways may be involved in depression-induced loss of gastric ICC.


Asunto(s)
Depresión/metabolismo , Mucosa Gástrica/metabolismo , Células Intersticiales de Cajal/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo , Animales , Peso Corporal , Proliferación Celular , Depresión/fisiopatología , Regulación hacia Abajo , Regulación de la Expresión Génica , Masculino , Miocitos del Músculo Liso/citología , Péptidos Natriuréticos/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores del Factor Natriurético Atrial/biosíntesis , Receptores del Factor Natriurético Atrial/metabolismo , Factor de Células Madre/biosíntesis , Sales de Tetrazolio/química , Tiazoles/química , Regulación hacia Arriba
14.
Protein Expr Purif ; 120: 99-105, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26724416

RESUMEN

We here present a new method for the expression and purification of recombinant human stem cell factor (rhSCF(164)) in endotoxin-free ClearColi(®) BL21(DE3) cells harboring codon-optimized Profinity eXact™-tagged hSCF cDNA. Previously, we demonstrated that co-expression with thioredoxin increased the solubility of rhSCF in Escherichia coli BL21(DE3), and addition of l-arginine enhanced chromatography performance by removing the endotoxin-masked surface of rhSCF. Initially, we tried to express rhSCF in an endotoxin-free strain using a thioredoxin co-expression system, which resulted in significantly lower expression, possibly due to the stress imposed by overexpressed thioredoxin or antibiotics susceptibility. Therefore, we developed a new expression system without thioredoxin. External redox coupling was tested using persulfides such as glutathione persulfide or cysteine persulfide for the in vivo-folding of hSCF in the cytoplasm. Persulfides improved the protein solubility by accelerating disulfide-exchange reactions for incorrectdisulfides during folding in E. coli. Furthermore, the persulfides enhanced the expression level, likely due to upregulation of the enzymatic activity of T7 RNA polymerase. The recombinant protein was purified via affinity chromatography followed by cleavage with sodium fluoride, resulting in complete proteolytic removal of the N-terminal tag. The endotoxin-free fusion protein from ClearColi(®) BL21(DE3) could bind to the resin in the standard protocol using sodium phosphate (pH 7.2). Furthermore, purified rhSCF enhanced the proliferation and maturation of the human mast cell line LAD2. Thus, we conclude that use of the protein expression system employing E. coli by disulfide shuffling with persulfide addition could be a very useful method for efficient protein production.


Asunto(s)
Cisteína/análogos & derivados , Disulfuros/metabolismo , Escherichia coli/genética , Glutatión/análogos & derivados , Factor de Células Madre/genética , Cromatografía de Afinidad , Clonación Molecular , Cisteína/metabolismo , Glutatión/metabolismo , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Solubilidad , Factor de Células Madre/biosíntesis , Factor de Células Madre/aislamiento & purificación , Regulación hacia Arriba
15.
J Clin Invest ; 125(10): 3965-80, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26389678

RESUMEN

Erythropoiesis is an important response to certain types of stress, including hypoxia, hemorrhage, bone marrow suppression, and anemia, that result in inadequate tissue oxygenation. This stress-induced erythropoiesis is distinct from basal red blood cell generation; however, neither the cellular nor the molecular factors that regulate this process are fully understood. Here, we report that type 1 conventional dendritic cells (cDC1s), which are defined by expression of CD8α in the mouse and XCR1 and CLEC9 in humans, are critical for induction of erythropoiesis in response to stress. Specifically, using murine models, we determined that engagement of a stress sensor, CD24, on cDC1s upregulates expression of the Kit ligand stem cell factor on these cells. The increased expression of stem cell factor resulted in Kit-mediated proliferative expansion of early erythroid progenitors and, ultimately, transient reticulocytosis in the circulation. Moreover, this stress response was triggered in part by alarmin recognition and was blunted in CD24 sensor- and CD8α+ DC-deficient animals. The contribution of the cDC1 subset to the initiation of stress erythropoiesis was distinct from the well-recognized role of macrophages in supporting late erythroid maturation. Together, these findings offer insight into the mechanism of stress erythropoiesis and into disorders of erythrocyte generation associated with stress.


Asunto(s)
Células Dendríticas/fisiología , Eritropoyesis/fisiología , Estrés Fisiológico/fisiología , Alarminas/fisiología , Animales , Antígeno CD24/fisiología , Antígenos CD8/análisis , Cisplatino/toxicidad , Ensayo de Unidades Formadoras de Colonias , Células Dendríticas/clasificación , Células Precursoras Eritroides/fisiología , Femenino , Perfilación de la Expresión Génica , Proteína HMGB1/toxicidad , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Hipoxia/fisiopatología , Mesilato de Imatinib/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Flebotomía/efectos adversos , Quimera por Radiación , Proteínas Recombinantes/toxicidad , Esplenectomía/efectos adversos , Factor de Células Madre/biosíntesis , Factor de Células Madre/genética
16.
Cell Signal ; 27(11): 2241-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26232617

RESUMEN

High glucose-insulted bone marrow-derived mesenchymal stem cells (BMCs) showed impaired angiogenesis along with downregulation of stem cell factor (SCF). This study was designed to determine the involvement of microRNAs (miR), which are actively involved in the physiological function of stem cells. We observed that miR-34c was significantly induced by high glucose treatment and blunted tube formation of BMCs. Stem cell factor (SCF) was confirmed as a target of miR-34c by 3'-UTR promoter analysis and Western blot. SCF knockdown by siRNA induced Krüppel-like factor 4 (KLF4) and resulted in the blockade of angiogenesis of BMCs. Sequentially, KLF4 overexpression completely blocked tube formation through inducing PAI-1 (plasminogen activator inhibitor-1). To study the action of miR-34c in terms of the therapeutic potential of BMCs, myocardial infarction (MI) was induced by ligation of the coronary artery in nude mice, BMCs transfected with miR-control or miR-34c were injected into the infarcted myocardium 7 days later, and histological studies were performed 2 weeks later. Cardiac fibrosis was 18.24±4.7% in the miR-34c-BMC group and 10.01±0.2% in the miR-control-BMC group (p<0.05). Cardiac function and vessel density were decreased in the miR-34c-BMC group compared with the miR-con-BMC group. Particularly, miR-34c-BMCs failed to incorporate into vessels. Our results show that the angiogenic activity of BMCs is finely regulated by the miR-34c-SCF-KLF4 axis, which is a potent translational target for optimizing the therapeutic activity of autologous BMCs for cardiac repair.


Asunto(s)
Glucosa/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/genética , Infarto del Miocardio/terapia , Factor de Células Madre/genética , Animales , Células de la Médula Ósea/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Vasos Coronarios/cirugía , Regulación hacia Abajo , Fibrosis/patología , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/biosíntesis , Infarto del Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factor de Células Madre/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Iran Biomed J ; 19(4): 220-5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26175108

RESUMEN

BACKGROUND: Ovarian tissue cryopreservation is an alternative strategy to preserve the fertility of women predicted to undergo premature ovarian failure. This study was designed to evaluate the expression of folliculogenesis-related genes, including factor in the germline alpha (FIGLA), growth differentiation factor-9 (GDF-9), follicle-stimulating hormone receptor (FSHR), and KIT LIGAND after vitrification/warming of human ovarian tissue. METHODS: Human ovarian tissue samples were collected from five transsexual women. In the laboratory, the ovarian medullary part was removed by a surgical blade, and the cortical tissue was cut into small pieces. Some pieces were vitrified and warmed and the others were considered as non-vitrified group (control). Follicular normality was assessed with morphological observation by a light microscope, and the expression of FIGLA, KIT LIGAND, GDF-9,, and FSHR genes was examined using real-time RT-PCR in both the vitrified and non-vitrified groups. RESULTS: Overall, 85% of the follicles preserved their normal morphologic feature after warming. The percentage of normal follicles and the expression of FIGLA, KIT LIGAND, GDF-9, and FSHR genes were similar in both vitrified and non-vitrified groups (P > 0.05). CONCLUSION: Vitrification/warming of human ovarian tissue had no remarkable effect on the expression of folliculogenesis-related genes.


Asunto(s)
Criopreservación/métodos , Preservación de la Fertilidad/métodos , Folículo Ovárico/citología , Folículo Ovárico/fisiología , Vitrificación , Adulto , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Femenino , Regulación de la Expresión Génica , Factor 9 de Diferenciación de Crecimiento/biosíntesis , Factor 9 de Diferenciación de Crecimiento/genética , Humanos , Masculino , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Factor de Células Madre/biosíntesis , Factor de Células Madre/genética , Técnicas de Cultivo de Tejidos , Personas Transgénero , Adulto Joven
18.
Sci Rep ; 5: 11410, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-26090649

RESUMEN

Lysophosphatidic acid (LPA) is a pleiotropic lipid mediator that acts through G protein-coupled receptors (LPA1-6). Although several biological roles of LPA4 are becoming apparent, its role in hematopoiesis has remained unknown. Here, we show a novel regulatory role for LPA4 in hematopoiesis. Lpar4 mRNA was predominantly expressed in mouse bone marrow (BM) PDGFRα(+) stromal cells, known as the components of the hematopoietic stem/progenitor cell (HSPC) niche. Compared with wild-type mice, LPA4-deficient mice had reduced HSPC numbers in the BM and spleen and were hypersusceptible to myelosuppression, most likely due to impairments in HSPC recovery and stem cell factor production in the BM. Analysis of reciprocal BM chimeras (LPA4-deficient BM into wild-type recipients and vice versa) indicated that stromal cells likely account for these phenotypes. Consistently, LPA4-deficient BM stromal cells showed downregulated mRNA expression of stem cell factor and tenascin-c in vitro. Taken together, these results suggest a critical and novel role for the LPA/LPA4 axis in regulating BM stromal cells.


Asunto(s)
Hematopoyesis , Células Madre Mesenquimatosas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores Purinérgicos/metabolismo , Animales , Antígenos de Superficie/metabolismo , Biomarcadores , Médula Ósea , Células de la Médula Ósea/metabolismo , Recuento de Células , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacología , Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Noqueados , Modelos Animales , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Receptores Purinérgicos/genética , Bazo , Factor de Células Madre/biosíntesis
19.
Wound Repair Regen ; 23(4): 583-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26032674

RESUMEN

Diabetic skin wounds lack the ability to heal properly and constitute a major and significant complication of diabetes. Nontraumatic lower extremity amputations are the number one complication of diabetic skin wounds. The complexity of their pathophysiology requires an intervention at many levels to enhance healing and wound closure. Stem cells are a promising treatment for diabetic skin wounds as they have the ability to correct abnormal healing. Stem cell factor (SCF), a chemokine expressed in the skin, can induce stem cells migration, however the role of SCF in diabetic skin wound healing is still unknown. We hypothesize that SCF would correct the impairment and promote the healing of diabetic skin wounds. Our results show that SCF improved wound closure in diabetic mice and increased HIF-1α and vascular endothelial growth factor (VEGF) expression levels in these wounds. SCF treatment also enhanced the migration of red fluorescent protein (RFP)-labeled skin stem cells via in utero intra-amniotic injection of lenti-RFP at E8. Interestingly these RFP+ cells are present in the epidermis, stain negative for K15, and appear to be distinct from the already known hair follicle stem cells. These results demonstrate that SCF improves diabetic wound healing in part by increasing the recruitment of a unique stem cell population present in the skin.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Preñez , Piel/lesiones , Factor de Células Madre/genética , Células Madre/patología , Cicatrización de Heridas/genética , Heridas y Lesiones/genética , Animales , Animales Recién Nacidos , Movimiento Celular/fisiología , Diabetes Mellitus Experimental , Femenino , Inmunohistoquímica , Queratina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Embarazo , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/embriología , Piel/metabolismo , Factor de Células Madre/biosíntesis , Células Madre/metabolismo , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
20.
J Ovarian Res ; 8: 31, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-26008799

RESUMEN

BACKGROUND: The c-kit/kit ligand (KITL) signalling axis is an essential component of ovarian folliculogenesis in mammals, but little is known about expression and localisation of its key components in the ovaries of reproductive age women. This study aimed to characterise mRNA expression of c-kit and KITL isoforms and the localisation of c-kit and KITL proteins in adult human premenopausal ovaries. METHODS: This study utilised granulosa cells obtained from the preovulatory follicles of women undergoing assisted reproduction, pieces of ovarian tissue obtained from premenopausal women undergoing gynaecological surgeries and archival paraffin-embedded premenopausal ovarian tissues. Methodology included PCR for gene expression and Western blot or immunohistochemistry for protein expression. RESULTS: Both c-kit mRNA isoforms, known as GNNK+ and GNNK-, were detected in human ovarian cortex, while KITL protein isoforms (KITL1 and KITL2) were present in ovarian cortex and human granulosa cells. Immunohistochemistry showed expression of KITL and c-kit protein in multiple cell types within follicles throughout development, from primordial follicles to large antral follicles, in addition to atretic follicles. Oocytes of all follicle stages expressed c-kit protein exclusively. Interestingly, unlike animal models, expression of both proteins displayed a less cell-type specific distribution with immunostaining present in granulosa, theca and stromal cells, suggesting that autocrine signalling occurs within the human ovary. CONCLUSION: The results of this study indicate that c-kit/KITL signalling also occurs in the human ovary, as established in various animal models, and may involve previously unknown autocrine signalling.


Asunto(s)
Ovario/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , ARN Mensajero/biosíntesis , Factor de Células Madre/biosíntesis , Adulto , Comunicación Autocrina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células de la Granulosa/metabolismo , Humanos , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Ovario/crecimiento & desarrollo , ARN Mensajero/genética , Factor de Células Madre/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...