Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.719
Filtrar
1.
BMC Cancer ; 23(1): 251, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36922758

RESUMEN

BACKGROUND: Currently there are no established fertility preservation options for pre-pubertal boys facing cancer treatment. Granulocyte-colony stimulating factor (G-CSF) treatment has been proposed to be chemoprotective against spermatogonial cell loss in an alkylating chemotherapy model of busulfan treated adult mice. Having previously shown that exposure to the alkylating-like chemotherapy cisplatin resulted in a reduction in germ cell numbers in immature human testicular tissues, we here investigate whether G-CSF would prevent cisplatin-induced germ cell loss in immature human and mouse (fetal and pre-pubertal) testicular tissues. METHODS: Organotypic in vitro culture systems were utilised to determine the effects of clinically-relevant concentrations of G-CSF in cisplatin-exposed immature testicular tissues. Human fetal (n = 14 fetuses) and mouse pre-pubertal (n = 4 litters) testicular tissue pieces were cultured and exposed to cisplatin or vehicle control for 24 hrs and analysed at 72 and 240 hrs post-exposure. Combined G-CSF and cisplatin exposure groups explored varying concentrations and duration of G-CSF supplementation to the culture medium (including groups receiving G-CSF before, during and after cisplatin exposure). In addition, effects of G-CSF supplementation alone were investigated. Survival of total germ cell and sub-populations were identified by expression of AP2γ and MAGE-A4 for human gonocytes and (pre)spermatogonia, respectively, and MVH and PLZF, for mouse germ cells and putative spermatogonial stem cells (SSCs) respectively, were quantified. RESULTS: Exposure to cisplatin resulted in a reduced germ cell number in human fetal and mouse pre-pubertal testicular tissues at 240 hrs post-exposure. Germ cell number was not preserved by combined exposure with G-CSF using any of the exposure regimens (prior to, during or after cisplatin exposure). Continuous supplementation with G-CSF alone for 14 days did not change the germ cell composition in either human or mouse immature testicular tissues. CONCLUSIONS: This study demonstrates that exposure to G-CSF does not prevent cisplatin-induced germ cell loss in immature human and mouse testicular tissues in an in vitro system.


Asunto(s)
Cisplatino , Testículo , Masculino , Humanos , Animales , Ratones , Testículo/metabolismo , Cisplatino/farmacología , Espermatogonias , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factores Estimulantes de Colonias/metabolismo , Factores Estimulantes de Colonias/farmacología , Granulocitos
2.
Transplant Proc ; 54(6): 1615-1617, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35914968

RESUMEN

In simultaneous pancreas-kidney transplantation (SPK) recipients, cytomegalovirus (CMV) infection is a major complication that has been associated with the use of tacrolimus (TAC)-based immunosuppression. As one of the immunosuppressive drug options, the use of mammalian target of rapamycin inhibitors (mTORi) results in reduced rates of CMV infection in the field of solid organ transplantation. However, little is known about mTORi usage in pancreas transplantation. We report a case of recurrent CMV infection that was controlled by the introduction of mTORi (everolimus) in addition to a TAC-based immunosuppression regimen in SPK. A 52-year-old man underwent SPK. Graft duodenal perforation occurred on the 13th day of surgery, and graft duodenal resection was performed after long-term abscess drainage treatment. After graft duodenal resection, he was diagnosed with CMV viremia, and valganciclovir was started. However, because of recurrent febrile neutropenia caused by cytopenia as a side effect of valganciclovir, there was a repeated need for granulocyte-colony stimulating factor treatment. Immunosuppressive drug taper adjustment was attempted to control recurrent CMV viremia, and everolimus was introduced with the aim of reducing the dose of TAC and mycophenolate mofetil. This resulted in a continuously negative CMV antigenemia test and a stable general condition. Understanding the characteristics of various immunosuppressive agents and appropriately controlling and managing infectious diseases is crucial for the good postoperative management of patients with SPK.


Asunto(s)
Infecciones por Citomegalovirus , Trasplante de Riñón , Trasplante de Páncreas , Factores Estimulantes de Colonias/farmacología , Factores Estimulantes de Colonias/uso terapéutico , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/tratamiento farmacológico , Everolimus/efectos adversos , Rechazo de Injerto/tratamiento farmacológico , Supervivencia de Injerto , Humanos , Inmunosupresores/efectos adversos , Trasplante de Riñón/efectos adversos , Masculino , Persona de Mediana Edad , Ácido Micofenólico/uso terapéutico , Páncreas , Trasplante de Páncreas/efectos adversos , Trasplante de Páncreas/métodos , Serina-Treonina Quinasas TOR , Tacrolimus/uso terapéutico , Valganciclovir/uso terapéutico , Viremia/tratamiento farmacológico
3.
Cell Mol Life Sci ; 79(4): 219, 2022 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-35366105

RESUMEN

Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Factores Estimulantes de Colonias/farmacología , Factores Estimulantes de Colonias/uso terapéutico , Microglía/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico
4.
Mini Rev Med Chem ; 21(13): 1638-1645, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390132

RESUMEN

Pluripotent stem cells of the bone marrow are stimulated by different cytokines to proliferation and differentiation into various types of blood cells. These cytokines are mostly glycoproteins. Erythropoietin stimulates stem cells to the formation of erythrocytes while colony-stimulating factors cause the formation of different types of white blood cells. Stem cell factors play an important role in the maintenance and survival of blood cells of all types. Thrombopoietin stimulates stem cells to proliferation and formation of blood platelets. Granulocyte colony-stimulating factor is probably the most important drug in use. It stimulates stem cells to the formation of neutrophile granulocytes. It is often used in recombinant forms such as filgrastim in the treatment of neutropenia in cancer chemotherapy or AIDS. Its pegylated conjugates such as pegfilgrastim are also available. Its activity can be supported by plerixafor, a small molecule - bicyclam derivative acting as an indirect agonist of stem cells factor. It acts as an antagonist of CXCR4 receptor activation of which brakes hematopoiesis. The treatment of conditions accompanied by thrombocytopenia such as idiopathic thrombocytopenic purpura is currently not performed by thrombopoietin but synthetic agonists of its receptor are preferred. Romiplostim is a peptibody. It consists of a protein part interacting with the thrombopoietin receptor which is, however, different from thrombopoietin, and of Fc fragment of immunoglobulin G1. In contrast, small molecule thrombopoietin receptor agonists represented by eltrombopag can be given orally unlike all of the above.


Asunto(s)
Factores Estimulantes de Colonias/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Células Madre/farmacología , Trombopoyesis/efectos de los fármacos , Benzoatos/química , Benzoatos/farmacología , Plaquetas/citología , Plaquetas/metabolismo , Diferenciación Celular/efectos de los fármacos , Humanos , Hidrazinas/química , Hidrazinas/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Pirazoles/química , Pirazoles/farmacología , Receptores de Trombopoyetina/agonistas , Receptores de Trombopoyetina/metabolismo , Bibliotecas de Moléculas Pequeñas/química
5.
Elife ; 92020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33236983

RESUMEN

Neutrophils constitute the largest population of phagocytic granulocytes in the blood of mammals. The development and function of neutrophils and monocytes is primarily governed by the granulocyte colony-stimulating factor receptor family (CSF3R/CSF3) and macrophage colony-stimulating factor receptor family (CSF1R/IL34/CSF1) respectively. Using various techniques this study considered how the emergence of receptor:ligand pairings shaped the distribution of blood myeloid cell populations. Comparative gene analysis supported the ancestral pairings of CSF1R/IL34 and CSF3R/CSF3, and the emergence of CSF1 later in lineages after the advent of Jawed/Jawless fish. Further analysis suggested that the emergence of CSF3 lead to reorganisation of granulocyte distribution between amphibian and early reptiles. However, the advent of endothermy likely contributed to the dominance of the neutrophil/heterophil in modern-day mammals and birds. In summary, we show that the emergence of CSF3R/CSF3 was a key factor in the subsequent evolution of the modern-day mammalian neutrophil.


Asunto(s)
Evolución Biológica , Cordados/fisiología , Factores Estimulantes de Colonias/farmacología , Simulación por Computador , Células Mieloides/fisiología , Neutrófilos/fisiología , Animales , Cordados/genética , Regulación de la Expresión Génica/fisiología , Especificidad de la Especie
6.
Immunol Lett ; 212: 6-13, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31154052

RESUMEN

In the last decade several studies employing stem cells-based therapies have been investigated as an optional treatment for multiple sclerosis. Several preclinical and few clinical studies tested the efficacy of mesenchymal stem cells as a potent candidate for such therapies. Here we suggest the option of "neuralization" of classical mesenchymal stem cells as a cellular structure that resembles neural stem cells as well as there differentiation by a unique procedure towards terminally differentiated neural cells suggesting that this cell population may be appropriate for clinical application in the CNS. We investigated whether neuralized MSC (NMSC) could promote repair and recovery after injection into mice with EAE. Injection of NMSC and differentiated NMSC starting at the onset of the chronic phase of disease improved neurological function compared to controls as well as compared to naïve MSC. Injection of NMSC and mainly differentiated correlated with a reduction in the inflammation as well as in the axonal loss/damage and reduced area of demyelination. These observations suggest that NMSC and differentiated NMSC may suggest a more potent cell-based therapy that naïve MSC in the treatment arsenal of multiple sclerosis.


Asunto(s)
Transdiferenciación Celular/inmunología , Factores Estimulantes de Colonias/farmacología , Encefalomielitis Autoinmune Experimental/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Esclerosis Múltiple/terapia , Animales , Técnicas de Cultivo de Célula , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Ratones , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Células-Madre Neurales/inmunología , Esferoides Celulares , Resultado del Tratamiento
7.
J Natl Compr Canc Netw ; 17(5.5): 622-624, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31117042

RESUMEN

Biosimilars are here to stay, but whether they will enjoy widespread uptake remains to be seen. The FDA sets a high bar for approval of biosimilar products, yet many clinicians remain skeptical about the efficacy and safety of these agents. Favorable experience with >30 biosimilars in Europe provides some reassurance that these agents are safe and effective and can be substituted for the reference product.


Asunto(s)
Anemia/tratamiento farmacológico , Biosimilares Farmacéuticos/uso terapéutico , Factores Estimulantes de Colonias/uso terapéutico , Aprobación de Drogas , Anemia/diagnóstico , Anemia/etiología , Biosimilares Farmacéuticos/farmacología , Factores Estimulantes de Colonias/farmacología , Manejo de la Enfermedad , Aprobación de Drogas/métodos , Humanos , Estados Unidos
8.
J Dairy Sci ; 102(4): 3439-3451, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30799104

RESUMEN

The objective of this study was to evaluate the effect of pegbovigrastim (PEG) treatment of peripartum Holstein cows on the microbiome found in the vagina postpartum using sequencing of the 16S rRNA gene. A subset of cows was randomly sampled from a larger study where cows had been randomly assigned to 1 of 2 treatments: pegbovigrastim (PEG) or untreated control (CTR). The PEG-treated cows received a subcutaneous injection containing 15 mg of pegbovigrastim 7 d before expected calving and a second injection within 24 h of calving. Vaginal samples from 97 PEG-treated and 98 CTR cows were collected at calving, 7 ± 3, and 35 ± 3 d in milk (0, 7, and 35 DIM). Metritis was diagnosed at 7 ± 3 DIM and purulent vaginal discharge (PVD) at 35 ± 3 DIM. The PEG treatment did not alter the vaginal microbiome. Principal coordinate analysis (PCoA) showed that metritic cows had a dissimilar vaginal microbiome compared with cows that did not develop metritis, particularly at 7 but also at 35 DIM. This difference was characterized by higher relative abundance of Porphyromonas and Bacteroides and a lower relative abundance of Ureaplasma, Ruminococcaceae, and Clostridiales at 7 DIM, and a higher relative abundance of Ureaplasma and a lower relative abundance of Pasteurellaceae at 35 DIM. Based on PCoA, we observed that cows that developed PVD had a dissimilar vaginal microbiome compared with cows that did not develop PVD, particularly at 35 DIM but also at 7 DIM. This difference was characterized by a higher relative abundance of Bacteroides at 7 DIM and higher relative abundance of Fusobacterium and Bacteroides at 35 DIM. Cows that developed metritis and PVD also had higher relative abundance of Fusobacterium and Bacteroides at 0 DIM. Furthermore, the Chao1 and Shannon indices were decreased in metritic cows at 7 DIM and in PVD cows at 7 and 35 DIM. In summary, PEG treatment had no effect on the vaginal microbiome, and uterine disease was associated with major changes in the microbiome found in the vagina postpartum.


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Factores Estimulantes de Colonias/farmacología , Endometritis/veterinaria , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Microbiota/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Vagina/microbiología , Animales , Bacteroides , Bovinos , Enfermedades de los Bovinos/microbiología , Endometritis/prevención & control , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Periodo Posparto , ARN Ribosómico 16S , Distribución Aleatoria , Proteínas Recombinantes/farmacología , Enfermedades Uterinas
9.
Cytokine ; 122: 154190, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-29074034

RESUMEN

Colony stimulating factors (CSFs) play a central role in the development and functional maturation of immune cells besides having pleiotropic effects on cells of the vascular wall. The production of CSFs is induced by multiple atherogenic and inflammatory stimuli and their expression levels are often correlated positively with advanced atherosclerotic plaques and adverse cardiovascular events in humans suggesting that CSFs play a critical role in the pathophysiology of atherosclerosis progression. Interestingly, recombinant CSFs as well as anti-CSFs are being increasingly used for diverse clinical indications. However, the effect of these novel therapeutics on atherosclerotic plaque progression is not well understood. Herein, we summarize the currently available literature on the complex role of CSFs in various stages of atherosclerosis and emphasize the necessity for conducting further mechanistic studies in animal models of atherosclerosis as well as the need for evaluating the cardiovascular safety of CSF-based therapies in humans.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/metabolismo , Animales , Aterosclerosis/metabolismo , Diferenciación Celular/efectos de los fármacos , Factores Estimulantes de Colonias/metabolismo , Factores Estimulantes de Colonias/farmacología , Progresión de la Enfermedad , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-3/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Modelos Animales , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/metabolismo
10.
EMBO J ; 36(22): 3336-3355, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29038174

RESUMEN

Accumulation of tumor-associated macrophages (TAMs) associates with malignant progression in cancer. However, the mechanisms that drive the pro-tumor functions of TAMs are not fully understood. ZEB1 is best known for driving an epithelial-to-mesenchymal transition (EMT) in cancer cells to promote tumor progression. However, a role for ZEB1 in macrophages and TAMs has not been studied. Here we describe that TAMs require ZEB1 for their tumor-promoting and chemotherapy resistance functions in a mouse model of ovarian cancer. Only TAMs that expressed full levels of Zeb1 accelerated tumor growth. Mechanistically, ZEB1 expression in TAMs induced their polarization toward an F4/80low pro-tumor phenotype, including direct activation of Ccr2 In turn, expression of ZEB1 by TAMs induced Ccl2, Cd74, and a mesenchymal/stem-like phenotype in cancer cells. In human ovarian carcinomas, TAM infiltration and CCR2 expression correlated with ZEB1 in tumor cells, where along with CCL2 and CD74 determined poorer prognosis. Importantly, ZEB1 in TAMs was a factor of poorer survival in human ovarian carcinomas. These data establish ZEB1 as a key factor in the tumor microenvironment and for maintaining TAMs' tumor-promoting functions.


Asunto(s)
Carcinogénesis/patología , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias/metabolismo , Neoplasias/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Antígenos de Diferenciación de Linfocitos B/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2/farmacología , Factores Estimulantes de Colonias/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Monocitos/efectos de los fármacos , Monocitos/patología , Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fenotipo , Receptores CCR2/metabolismo , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos
11.
Dev Cell ; 42(5): 498-513.e6, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28867487

RESUMEN

The Arp2/3 complex nucleates branched actin, forming networks involved in lamellipodial protrusion, phagocytosis, and cell adhesion. We derived primary bone marrow macrophages lacking Arp2/3 complex (Arpc2-/-) and directly tested its role in macrophage functions. Despite protrusion and actin assembly defects, Arpc2-/- macrophages competently phagocytose via FcR and chemotax toward CSF and CX3CL1. However, CR3 phagocytosis and fibronectin haptotaxis, both integrin-dependent processes, are disrupted. Integrin-responsive actin assembly and αM/ß2 integrin localization are compromised in Arpc2-/- cells. Using an in vivo system to observe endogenous monocytes migrating toward full-thickness ear wounds we found that Arpc2-/- monocytes maintain cell speeds and directionality similar to control. Our work reveals that the Arp2/3 complex is not a general requirement for phagocytosis or chemotaxis but is a critical driver of integrin-dependent processes. We demonstrate further that cells lacking Arp2/3 complex function in vivo remain capable of executing important physiological responses that require rapid directional motility.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Movimiento Celular , Integrinas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Fagocitosis , Receptores Fc/metabolismo , Actinas/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Quimiocina CX3CL1/farmacología , Quimiotaxis/efectos de los fármacos , Factores Estimulantes de Colonias/farmacología , Femenino , Fibronectinas/farmacología , Ligandos , Antígeno de Macrófago-1/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/ultraestructura , Masculino , Ratones Endogámicos C57BL , Cadenas Pesadas de Miosina/metabolismo , Fagocitosis/efectos de los fármacos , Fenotipo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Expert Opin Biol Ther ; 17(7): 861-870, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28429626

RESUMEN

INTRODUCTION: Despite the relatively high efficacy of antifungal drugs, invasive fungal infections (IFIs) are still associated with tremendous morbidity and mortality, since late diagnosis makes an antifungal drug therapy inefficient. Therefore, antifungal immunotherapies to specifically strengthen the host´s own immune mechanisms constitute an additional promising strategy in taking action against fungal pathogens. Areas covered: The authors summarize efforts in research and clinical trials to provide safe and efficient immunotherapeutic options against invasive fungal diseases. Treatment of IFIs is challenging as the number of available antifungals is limited and further complications include: toxicity, drug interactions and the emergence of drug resistance. Susceptibility is determined by the impaired immune status of the host. Hence, augmenting immunity by immunotherapeutic interventions may offer future directions to treat IFI. Expert opinion: A much better understanding of fungus and host cell interactions is essential for the development of safe and successful immunotherapeutic strategies. Indeed, there is encouraging preliminary data available that such approaches are possible; however, current data is too limited to allow solid conclusions on the risks and benefits in the clinical setting. Clinical trials focusing on the role of adjuvant immunotherapeutics with or without a combination of antifungals are highly needed for further evaluation.


Asunto(s)
Inmunoterapia , Micosis/terapia , Inmunidad Adaptativa , Factores Estimulantes de Colonias/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Vacunas Fúngicas/inmunología , Humanos , Inmunidad Innata , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Micosis/inmunología , Micosis/prevención & control , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología
13.
Taiwan J Obstet Gynecol ; 55(5): 738-740, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27751427

RESUMEN

OBJECTIVE: Neutropenia developed after continuous intravenous infusion of ritodrine hydrochloride (Yutopar) for preterm uterine contractions in a twin pregnancy. We successfully returned the low neutrophil count to the normal range after discontinuation of infusion of ritodrine and treatment with granulocyte colony stimulating factor (G-CSF). CASE REPORT: A 34-year-old woman with twin pregnancy was treated with ritodrine for preterm uterine contractions at 27 weeks and 6 days gestation. Neutropenia developed after continuous intravenous infusion of ritodrine for about 4 weeks. We ceased the ritodrine infusion immediately and treated the neutropenia with G-CSF. A cesarean delivery was performed the day after cessation of the ritodrine infusion because of uncontrolled preterm labor. There were no adverse side effects or infectious complications in the mother or the newborns. The maternal neutrophil count recovered to the normal range 4 days after administration of G-CSF. CONCLUSION: Based on prior case reports and the clinical presentation of our case, G-CSF may be a useful treatment for pregnant women with ritodrine-induced neutropenia. However, more clinical studies are required to confirm the safety and efficacy of this treatment.


Asunto(s)
Factores Estimulantes de Colonias/farmacología , Neutropenia/tratamiento farmacológico , Embarazo Gemelar , Ritodrina/efectos adversos , Adulto , Cesárea , Femenino , Humanos , Recuento de Leucocitos , Neutropenia/inducido químicamente , Neutropenia/diagnóstico , Embarazo , Tocolíticos/efectos adversos
14.
Reproduction ; 152(6): 645-654, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27601717

RESUMEN

We tested whether gene expression of the bovine morula is modified by CSF2 in a sex-dependent manner and if sex determines the effect of CSF2 on competence of embryos to become blastocysts. Embryos were produced in vitro using X- or Y-sorted semen and treated at Day 5 of culture with 10 ng/mL bovine CSF2 or control. In experiment 1, morulae were collected at Day 6 and biological replicates (n = 8) were evaluated for transcript abundance of 90 genes by RT-qPCR using the Fluidigm Delta Gene assay. Expression of more than one-third (33 of 90) of genes examined was affected by sex. The effect of CSF2 on gene expression was modified by sex (P < 0.05) for five genes (DDX3Y/DDX3X-like, NANOG, MYF6, POU5F1 and RIPK3) and tended (P < 0.10) to be modified by sex for five other genes (DAPK1, HOXA5, PPP2R3A, PTEN and TNFSF8). In experiment 2, embryos were treated at Day 5 with control or CSF2 and blastocysts were collected at Day 7 for immunolabeling to determine the number of inner cell mass (ICM) and trophectoderm (TE) cells. CSF2 increased the percent of putative zygotes that became blastocysts for females, but did not affect the development of males. There was no effect of CSF2 or interaction of CSF2 with sex on the total number of blastomeres in blastocysts or in the number of inner cell mass or trophectoderm cells. In conclusion, CSF2 exerted divergent responses on gene expression and development of female and male embryos. These results are evidence of sexually dimorphic responses of the preimplantation embryo to this embryokine.


Asunto(s)
Blastocisto/efectos de los fármacos , Factores Estimulantes de Colonias/farmacología , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Animales , Blastocisto/citología , Blastocisto/metabolismo , Bovinos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Linaje de la Célula , Técnicas de Cultivo de Embriones , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Masculino , Factores Sexuales , Transducción de Señal/efectos de los fármacos
15.
PLoS One ; 11(2): e0150230, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925951

RESUMEN

It is increasingly clear that nicotinic acetylcholine receptors (nAChRs) are involved in immune regulation, and that their activation can protect against inflammatory diseases. Previous data have shown that nicotine diminishes the numbers of peripheral monocytes and macrophages, especially those of the pro-inflammatory phenotype. The goal of the present study was to determine if nicotine modulates the production of bone marrow -derived monocytes/macrophages. In this study, we first found that murine bone marrow cells express multiple nAChR subunits, and that the α7 and α9 nAChRs most predominant subtypes found in immune cells and their precursors. Using primary cultures of murine bone marrow cells, we then determined the effect of nicotine on monocyte colony-stimulating factor and interferon gamma (IFNγ)-induced monocyte production. We found that nicotine lowered the overall number of monocytes, and more specifically, inhibited the IFNγ-induced increase in pro-inflammatory monocytes by reducing cell proliferation and viability. These data suggested that nicotine diminishes the ratio of pro-inflammatory versus anti-inflammatory monocyte produced in the bone marrow. We thus confirmed this hypothesis by measuring cytokine expression, where we found that nicotine inhibited the production of the pro-inflammatory cytokines TNFα, IL-1ß and IL-12, while stimulating the secretion of IL-10, an anti-inflammatory cytokine. Finally, nicotine also reduced the number of pro-inflammatory monocytes in the bone marrow of LPS-challenged mice. Overall, our data demonstrate that both α7 and α9 nAChRs are involved in the regulation of pro-inflammatory M1 monocyte numbers.


Asunto(s)
Monocitos/citología , Monocitos/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factores Estimulantes de Colonias/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/farmacología , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Nicotina/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Nicotínicos/genética
16.
Arch Dermatol Res ; 307(4): 341-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25666388

RESUMEN

Melanogenic paracrine and autocrine cytokine networks have recently been discovered in vitro between melanocytes and other types of skin cells. Granulocyte colony-stimulating factor receptor (CSF3R) controls the survival, proliferation and differentiation of many kinds of cells, including neutrophils. To understand the function of CSF3R and recombinant human granulocyte colony-stimulating factor (rhCSF3) on melanocyte proliferation, this study compared the expression of CSF3R and the effects of rhCSF3 in primary human melanocytes, neutrophils and HEL 92.1.7 cells. The results show that CSF3R is localized in the cytoplasm and on cell membranes of melanocytes and neutrophils. The percentage of CSF3R(+) melanocytes was higher than CSF3R(+) HEL 92.1.7 cells, but was lower than CSF3R(+) neutrophils. Both CSF3R mRNA and CSF3R protein levels in melanocytes were higher than in HEL 92.1.7 cells, but were lower than in neutrophils. Treatment with rhCSF3 increased the proliferation of human melanocytes, but not their tyrosinase activity. Transcripts of CSF3R in human melanocytes, M14, A375 melanoma and A431 squamous cell carcinoma cells were also detected. Expression of the CSF3R V3 transcript was lower in melanocytes than in M14, A375 melanoma and A431 squamous cell carcinoma cells. In conclusion, rhCSF3 can promote melanocyte proliferation through CSF3R without affecting tyrosinase activity.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Factores Estimulantes de Colonias/farmacología , Regulación de la Expresión Génica/fisiología , Melanocitos/citología , Melanocitos/metabolismo , Receptores del Factor Estimulante de Colonias/genética , Receptores del Factor Estimulante de Colonias/metabolismo , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Masculino , Monofenol Monooxigenasa/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , ARN Mensajero/genética , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
18.
Genet Mol Res ; 13(3): 6356-66, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24615120

RESUMEN

Colony stimulating factors (CSF) have been considered to modulate liver regeneration (LR) after partial hepatectomy (PH) at the tissue level. However, it remains unclear about precise mechanism of action of CSF in regeneration at the cellular level. Therefore, eight rat liver cell types were isolated by Percoll gradient centrifugation and magnetic beads. CSF-mediated signaling pathway genes were obtained by searching the related pathway databases and their expression profiles in 8 hepatic cell types were measured using rat Genome 230 2.0 Microarray. RT-PCR was performed to assess the reliability of chip results. The result showed a large difference in expression profiles of CSF-mediated signaling pathway genes between different cell types; most genes involved in CSF-mediated signaling pathways were mainly unregulated across liver cell samples. The implication of these genes in LR was analyzed by the bioinformatics and systems biology method. According to chip results and gene synergy, a significant enhancement of the CSF3-mediated Pi3k/Akt pathway at 30-36 h in hepatocytes and at 24 h in biliary epithelial cells post-PH could be associated with active proliferation in these two cell types; the striking decrease in Jak/Stat cascade activity in hepatic stellate cells at 2 and 12 h post- PH or even inactive in dendritric cells during the whole LR implied that proliferation of these two cell types is possibly regulated by other signaling pathways. These data suggest the potential relevance of CSF in liver regeneration at the cellular level.


Asunto(s)
Factores Estimulantes de Colonias/farmacología , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Regeneración Hepática/genética , Hígado/metabolismo , Animales , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Hepatectomía , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Macrófagos del Hígado/citología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/cirugía , Análisis por Micromatrices , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal
19.
Curr Cardiol Rev ; 10(1): 73-86, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23638829

RESUMEN

In the event of obstructive coronary artery disease, collateral arteries have been deemed an alternative blood source to preserve myocardial tissue perfusion and function. Monocytes play an important role in modulating this process, by local secretion of growth factors and extracellular matrix degrading enzymes. Extensive efforts have focused on developing compounds for augmenting the growth of collateral vessels (arteriogenesis). Nonetheless, clinical trials investigating the therapeutic potential of these compounds resulted in disappointing outcomes. Previous studies focused on developing compounds that stimulated collateral vessel growth by enhancing monocyte survival and activity. The limited success of these compounds in clinical studies, led to a paradigm shift in arteriogenesis research. Recent studies have shown genetic heterogeneity between CAD patients with sufficient and insufficient collateral vessels. The genetic predispositions in patients with poorly developed collateral vessels include overexpression of arteriogenesis inhibiting signaling pathways. New directions of arteriogenesis research focus on attempting to block such inhibitory pathways to ultimately promote arteriogenesis. Methods to detect collateral vessel growth are also critical in realizing the therapeutic potential of newly developed compounds. Traditional invasive measurements of intracoronary derived collateral flow index remain the gold standard in quantifying functional capacity of collateral vessels. However, advancements made in hybrid diagnostic imaging modalities will also prove to be advantageous in detecting the effects of pro-arteriogenic compounds.


Asunto(s)
Investigación Biomédica/tendencias , Circulación Colateral/fisiología , Enfermedad de la Arteria Coronaria/fisiopatología , Circulación Coronaria/fisiología , Oclusión Coronaria/fisiopatología , Inductores de la Angiogénesis/farmacología , Animales , Quimiocina CCL2/farmacología , Factores Estimulantes de Colonias/farmacología , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/diagnóstico , Oclusión Coronaria/diagnóstico , Oclusión Coronaria/terapia , Predicción , Humanos , Ratones , Monocitos/fisiología , Estrés Fisiológico/fisiología
20.
Br J Haematol ; 158(6): 778-87, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22816563

RESUMEN

In vitro exposure of haematopoietic stem and progenitor cells (HSPC) to cytokines in expansion or gene therapy protocols reduces homing and engraftment in vivo. We have previously reported that this is related in part to altered tissue specificity of short-term homing, leading to loss of cells in non-haematopoietic tissues. Here we demonstrate that defective engraftment persists when cultured HSPC are transplanted by intrabone injection. Changes in engraftment function occur within 24 h of cytokine exposure, and are evident when engraftment is analysed solely in the injected bone. A novel ex vivo model of the bone marrow was developed, in which the attachment of infused HSPC in rodent long bones is reduced following culture with cytokines. Finally, cultured HSPC demonstrated reduced adhesion to N-cadherin, osteopontin and vascular cell-adhesion molecule-1, ligands present in bone marrow niches. These changes in adhesive function occur rapidly, and are not related to downregulation of the relevant receptors. Our findings suggest that cytokine exposure of adult human HSPC results in altered adhesion within bone marrow niches, further leading to reduced engraftment potential in vivo.


Asunto(s)
Médula Ósea/patología , Adhesión Celular/fisiología , Factores Estimulantes de Colonias/farmacología , Supervivencia de Injerto/fisiología , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Nicho de Células Madre/fisiología , Adulto , Animales , Antígenos CD34/análisis , Células Cultivadas/efectos de los fármacos , Células Cultivadas/trasplante , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...