Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Curr Microbiol ; 81(8): 242, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38913141

RESUMEN

Chikungunya virus (CHIKV) is a causative agent of a disease continuum, ranging from an acute transient chikungunya fever to chronic incapacitating viral arthralgia. The interaction between anti-CHIKV antibodies and the complement system has recently received attention. However, the contribution of complement activation in CHIKV-induced pathologies has not been fully elucidated. The present study was undertaken to delineate the possible contribution of complement activation in CHIKV-induced disease progression. In this study, using plasma specimens of chikungunya patients in the acute, chronic, and recovered phases of infection, we explicated the involvement of complement activation in CHIKV disease progression by ELISAs and Bio-Plex assays. Correlation analysis was carried out to demonstrate interrelation among C1q-binding IgG-containing circulating immune complexes (CIC-C1q), complement activation fragments (C3a, C5a, sC5b-9), and complement-modulated pro-inflammatory cytokines (IL-1ß, IL-18, IL-6, and TNF-α). We detected elevated complement activation fragments, CIC-C1q, and complement-modulated cytokines in the varied patient groups compared with the healthy controls, indicating persistent activation of the complement system. Furthermore, we observed statistically significant correlations among CIC-C1q with complement activation fragments and C3a with complement modulatory cytokines IL-1ß, IL-6, and IL-18 during the CHIKV disease progression. Taken together, the current data provide insight into the plausible association between CICs, complement activation, subsequent complement modulatory cytokine expression, and CHIKV etiopathology.


Asunto(s)
Complejo Antígeno-Anticuerpo , Fiebre Chikungunya , Virus Chikungunya , Activación de Complemento , Complemento C1q , Citocinas , Humanos , Complemento C1q/metabolismo , Complemento C1q/inmunología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/virología , Fiebre Chikungunya/sangre , Complejo Antígeno-Anticuerpo/sangre , Complejo Antígeno-Anticuerpo/inmunología , Virus Chikungunya/inmunología , Masculino , Citocinas/sangre , Citocinas/metabolismo , Persona de Mediana Edad , Adulto , Femenino , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anciano , Adulto Joven
2.
Eur J Immunol ; 52(11): 1842-1858, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36074916

RESUMEN

IL-17-producing Th17 cells play an important role in pathogenesis of rheumatoid arthritis (RA). Aberrant immune activation due to an imbalance between Th17 and regulatory T (Treg) cells is associated with the development of RA and other autoimmune diseases. Targeting pathogenic Th17 cells and their associated molecules is emerging as a promising strategy to treat and reverse RA. Here, we demonstrate that IL-3 inhibits the differentiation of Th17 cells and promotes the development of Treg cells in IL-2-dependent manner. In IL-2 KO mice, we observed that IL-3 has no effect on differentiation of both Th17 and Treg cells. In addition, IL-3 decreases pathogenic IL-17A+ TNF-α+ , IL-17A+ IFN-γ+ and IL-23R+ Th17 cells, secretion of GM-CSF and IFN-γ, and osteoclastogenesis when presented in the culture together with Th17 polarizing cytokines. Mechanistically, IL-3 regulates the development of Th17 cells through the inhibition of STAT3 phosphorylation. IL-3 treatment significantly decreases the pathogenic Th17 cell responses and arthritic scores in the mouse model of RA. Importantly, IL-3 inhibits the differentiation of human Th17 cells. Thus, our results suggest a novel therapeutic role of IL-3 in the regulation of Th17 cell-mediated pathophysiology of RA.


Asunto(s)
Artritis Reumatoide , Diferenciación Celular , Interleucina-3 , Células Th17 , Animales , Humanos , Ratones , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-3/metabolismo , Linfocitos T Reguladores/citología , Células Th17/citología
3.
J Immunol ; 204(4): 819-831, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31900339

RESUMEN

IL-3, a cytokine secreted by activated T lymphocytes, is known to regulate the proliferation, survival, and differentiation of hematopoietic cells. However, the role of IL-3 in regulation of T cell functions is not fully delineated. Previously, we have reported that IL-3 plays an important role in development of regulatory T cells in mice. In this study, we investigated the regulation of IL-3R expression on human Th cells and also examined the role of IL-3 in effector functions of these cells. We found that human peripheral blood Th cells in resting state do not show surface expression of IL-3R; however, its expression was observed at transcript and intracellular protein levels. The functional IL-3R expression on the surface was seen only after antigenic stimulation. When naive Th cells were activated in the presence of various cytokines, we found that IL-4 significantly increases the surface expression of IL-3R and also increases the number of IL-3R+ Th cells. Interestingly, IL-3R+ cells exhibit a Th2 cell-like phenotype and show high GATA-3 expression. Moreover, Th2 cells in presence of IL-3 show increased expression of type 2 effector cytokines, such as IL-4, IL-5, and IL-13. Furthermore, IL-3R expressing and IL-3-secreting Th cells were high in house dust mite-allergic patients. Thus, to our knowledge, we provide the first evidence that the expression of IL-3R on activated human Th cells is modulated by IL-4, and IL-3 regulates the effector functions of Th2 cells. Our results suggest that IL-3 may play an important role in regulating allergic immune responses.


Asunto(s)
Diferenciación Celular/inmunología , Interleucina-3/inmunología , Interleucina-4/inmunología , Receptores de Interleucina-3/inmunología , Células Th2/inmunología , Humanos , Hipersensibilidad/inmunología , Interleucina-3/metabolismo , Interleucina-4/metabolismo , Activación de Linfocitos/inmunología , Receptores de Interleucina-3/metabolismo
4.
J Biol Chem ; 294(41): 14879-14895, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31413110

RESUMEN

F-box protein 31 (FBXO31) is a reported putative tumor suppressor, and its inactivation due to loss of heterozygosity is associated with cancers of different origins. An emerging body of literature has documented FBXO31's role in preserving genome integrity following DNA damage and in the cell cycle. However, knowledge regarding the role of FBXO31 during normal cell-cycle progression is restricted to its functions during the G2/M phase. Interestingly, FBXO31 levels remain high even during the early G1 phase, a crucial stage for preparing the cells for DNA replication. Therefore, we sought to investigate the functions of FBXO31 during the G1 phase of the cell cycle. Here, using flow cytometric, biochemical, and immunofluorescence techniques, we show that FBXO31 is essential for maintaining optimum expression of the cell-cycle protein cyclin A for efficient cell-cycle progression. Stable FBXO31 knockdown led to atypical accumulation of cyclin A during the G1 phase, driving premature DNA replication and compromised loading of the minichromosome maintenance complex, resulting in replication from fewer origins and DNA double-strand breaks. Because of these inherent defects in replication, FBXO31-knockdown cells were hypersensitive to replication stress-inducing agents and displayed pronounced genomic instability. Upon entering mitosis, the cells defective in DNA replication exhibited a delay in the prometaphase-to-metaphase transition and anaphase defects such as lagging and bridging chromosomes. In conclusion, our findings establish that FBXO31 plays a pivotal role in preserving genomic integrity by maintaining low cyclin A levels during the G1 phase for faithful genome duplication and segregation.


Asunto(s)
Ciclina A/metabolismo , Replicación del ADN/genética , Proteínas F-Box/metabolismo , Genoma Humano/genética , Proteínas Supresoras de Tumor/metabolismo , Ciclo Celular/genética , Cromatina/genética , Proteínas F-Box/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Cinética , Células MCF-7 , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Ubiquitinación/genética
5.
J Immunol ; 200(2): 595-606, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29203513

RESUMEN

Bone remodeling comprises balanced activities between osteoclasts and osteoblasts, which is regulated by various factors, including hormones and cytokines. We previously reported that IL-3 inhibits osteoclast differentiation and pathological bone loss. IL-3 also enhances osteoblast differentiation and bone formation from mesenchymal stem cells. However, the role of IL-3 in regulation of osteoblast-osteoclast interactions and underlying mechanisms is not yet delineated. In this study, we investigated the role of IL-3 on the regulation of osteoblast-specific molecules, receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG) that modulate bone homeostasis. We found that IL-3 increases RANKL expression at both the transcriptional and translational levels, and it showed no effect on OPG expression in calvarial osteoblasts. The increased RANKL expression by IL-3 induces mononuclear osteoclasts; however, it does not induce multinuclear osteoclasts. Interestingly, IL-3 decreases soluble RANKL by reducing ectodomain shedding of membrane RANKL through downregulation of metalloproteases mainly a disintegrin and metalloproteinase (ADAM)10, ADAM17, ADAM19, and MMP3. Moreover, IL-3 increases membrane RANKL by activating the JAK2/STAT5 pathway. Furthermore, IL-3 enhances RANKL expression in mesenchymal stem cells of wild-type mice but not in STAT5a knockout mice. Interestingly, IL-3 restores RANKL expression in adult mice by enhancing bone-specific RANKL and decreasing serum RANKL. Furthermore, IL-3 increases the serum OPG level in adult mice. Thus, our results reveal, to our knowledge for the first time, that IL-3 differentially regulates two functional forms of RANKL through metalloproteases and the JAK2/STAT5 pathway, and it helps in restoring the decreased RANKL/OPG ratio in adult mice. Notably, our studies indicate the novel role of IL-3 in regulating bone homeostasis in important skeletal disorders.


Asunto(s)
Interleucina-3/metabolismo , Janus Quinasa 2/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Osteoblastos/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Expresión Génica , Interleucina-3/farmacología , Ratones , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/sangre , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal/efectos de los fármacos
6.
EMBO J ; 33(9): 994-1010, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24431222

RESUMEN

There is emerging evidence that stem cells can rejuvenate damaged cells by mitochondrial transfer. Earlier studies show that epithelial mitochondrial dysfunction is critical in asthma pathogenesis. Here we show for the first time that Miro1, a mitochondrial Rho-GTPase, regulates intercellular mitochondrial movement from mesenchymal stem cells (MSC) to epithelial cells (EC). We demonstrate that overexpression of Miro1 in MSC (MSCmiro(Hi)) leads to enhanced mitochondrial transfer and rescue of epithelial injury, while Miro1 knockdown (MSCmiro(Lo)) leads to loss of efficacy. Treatment with MSCmiro(Hi) was associated with greater therapeutic efficacy, when compared to control MSC, in mouse models of rotenone (Rot) induced airway injury and allergic airway inflammation (AAI). Notably, airway hyperresponsiveness and remodeling were reversed by MSCmiro(Hi) in three separate allergen-induced asthma models. In a human in vitro system, MSCmiro(Hi) reversed mitochondrial dysfunction in bronchial epithelial cells treated with pro-inflammatory supernatant of IL-13-induced macrophages. Anti-inflammatory MSC products like NO, TGF-ß, IL-10 and PGE2, were unchanged by Miro1 overexpression, excluding non-specific paracrine effects. In summary, Miro1 overexpression leads to increased stem cell repair.


Asunto(s)
Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Mitocondrias/metabolismo , Proteínas de Unión al GTP rho/fisiología , Animales , Transporte Biológico/genética , Células Cultivadas , Terapia Genética/métodos , Humanos , Pulmón/patología , Lesión Pulmonar/patología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mitocondrias/trasplante , Células 3T3 NIH , Nanotubos , Resultado del Tratamiento , Proteínas de Unión al GTP rho/genética
7.
J Immunol ; 196(12): 5024-35, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183574

RESUMEN

Osteoarthritis (OA) is a chronic disease of articular joints that leads to degeneration of both cartilage and subchondral bone. These degenerative changes are further aggravated by proinflammatory cytokines including IL-1ß and TNF-α. Previously, we have reported that IL-3, a cytokine secreted by activated T cells, protects cartilage and bone damage in murine models of inflammatory and rheumatoid arthritis. However, how IL-3 protects cartilage degeneration is not yet known. In this study, we investigated the role of IL-3 on cartilage degeneration under both in vitro and in vivo conditions. We found that both mouse and human chondrocytes show strong expression of IL-3R at gene and protein levels. IL-3 increases the expression of mouse chondrocyte-specific genes, Sox9 and collagen type IIa, which were downregulated by IL-1ß. Moreover, IL-3 downregulated IL-1ß- and TNF-α-induced expression of matrix metalloproteinases in both mouse and human chondrocytes. Interestingly, IL-3 reduces the degeneration of articular cartilage and subchondral bone microarchitecture in a mouse model of human OA. Moreover, IL-3 showed the preventive and therapeutic effects on cartilage degeneration induced by IL-1ß in micromass pellet cultures of human mesenchymal stem cells. Thus, to our knowledge, we provide the first evidence that IL-3 has therapeutic potential in amelioration of degeneration of articular cartilage and subchondral bone microarchitecture associated with OA.


Asunto(s)
Cartílago Articular/patología , Regulación hacia Abajo , Interleucina-3/uso terapéutico , Metaloproteinasas de la Matriz/genética , Osteoartritis/tratamiento farmacológico , Animales , Cartílago Articular/efectos de los fármacos , Cartílago Articular/inmunología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/inmunología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/farmacología , Interleucina-3/administración & dosificación , Interleucina-3/farmacología , Subunidad alfa del Receptor de Interleucina-3/genética , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Ratones , Osteoartritis/inmunología , Osteoartritis/fisiopatología , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
J Immunol ; 195(11): 5136-48, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26538398

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammatory synovitis leading to joint destruction and systemic bone loss. The inflammation-induced bone loss is mediated by increased osteoclast formation and function. Current antirheumatic therapies primarily target suppression of inflammatory cascade with limited or no success in controlling progression of bone destruction. Mesenchymal stem cells (MSCs) by virtue of their tissue repair and immunomodulatory properties have shown promising results in various autoimmune and degenerative diseases. However, the role of MSCs in prevention of bone destruction in RA is not yet understood. In this study, we investigated the effect of adipose-derived MSCs (ASCs) on in vitro formation of bone-resorbing osteoclasts and pathological bone loss in the mouse collagen-induced arthritis (CIA) model of RA. We observed that ASCs significantly inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis in both a contact-dependent and -independent manner. Additionally, ASCs inhibited RANKL-induced osteoclastogenesis in the presence of proinflammatory cytokines such as TNF-α, IL-17, and IL-1ß. Furthermore, treatment with ASCs at the onset of CIA significantly reduced clinical symptoms and joint pathology. Interestingly, ASCs protected periarticular and systemic bone loss in CIA mice by maintaining trabecular bone structure. We further observed that treatment with ASCs reduced osteoclast precursors in bone marrow, resulting in decreased osteoclastogenesis. Moreover, ASCs suppressed autoimmune T cell responses and increased the percentages of peripheral regulatory T and B cells. Thus, we provide strong evidence that ASCs ameliorate inflammation-induced systemic bone loss in CIA mice by reducing osteoclast precursors and promoting immune tolerance.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Resorción Ósea/inmunología , Células Madre Mesenquimatosas/inmunología , Osteoclastos/inmunología , Ligando RANK/antagonistas & inhibidores , Tejido Adiposo/citología , Animales , Artritis Experimental/patología , Artritis Reumatoide/patología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Huesos/inmunología , Huesos/patología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Tolerancia Inmunológica/inmunología , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos DBA , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Biochem Biophys Res Commun ; 455(1-2): 133-8, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25450704

RESUMEN

The relationship between obesity and bone is complex. Epidemiological studies demonstrate positive as well as negative correlation between obesity and bone health. In the present study, we investigated the impact of high fat diet-induced obesity on peak bone mass. After 9 months of feeding young rats with high fat diet, we observed obesity phenotype in rats with increased body weight, fat mass, serum triglycerides and cholesterol. There were significant increases in serum total alkaline phosphatase, bone mineral density and bone mineral content. By micro-computed tomography (µ-CT), we observed a trend of better trabecular bones with respect to their microarchitecture and geometry. This indicated that high fat diet helps in achieving peak bone mass and microstructure at younger age. We subsequently shifted rats from high fat diet to normal diet for 6 months and evaluated bone/obesity parameters. It was observed that after shifting rats from high fat diet to normal diet, fat mass, serum triglycerides and cholesterol were significantly decreased. Interestingly, the gain in bone mineral density, bone mineral content and trabecular bone parameters by HFD was retained even after body weight and obesity were normalized. These results suggest that fat rich diet during growth could accelerate achievement of peak bone mass that is sustainable even after withdrawal of high fat diet.


Asunto(s)
Densidad Ósea , Dieta Alta en Grasa , Obesidad/etiología , Animales , Huesos/diagnóstico por imagen , Dieta Alta en Grasa/efectos adversos , Masculino , Radiografía , Ratas , Ratas Wistar
10.
J Oral Implantol ; 40 Spec No: 347-55, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25020216

RESUMEN

This investigation was carried out to study the effect of a novel process of surface modification, surface nanostructuring by ultrasonic shot peening, on osteoblast proliferation and corrosion behavior of commercially pure titanium (c p-Ti) in simulated body fluid. A mechanically polished disc of c p-Ti was subjected to ultrasonic shot peening with stainless steel balls to create nanostructure at the surface. A nanostructure (<20 nm) with inhomogeneous distribution was revealed by atomic force and scanning electron microscopy. There was an increase of approximately 10% in cell proliferation, but there was drastic fall in corrosion resistance. Corrosion rate was increased by 327% in the shot peened condition. In order to examine the role of residual stresses associated with the shot peened surface on these aspects, a part of the shot peened specimen was annealed at 400°C for 1 hour. A marked influence of annealing treatment was observed on surface structure, cell proliferation, and corrosion resistance. Surface nanostructure was much more prominent, with increased number density and sharper grain boundaries; cell proliferation was enhanced to approximately 50% and corrosion rate was reduced by 86.2% and 41% as compared with that of the shot peened and the as received conditions, respectively. The highly significant improvement in cell proliferation, resulting from annealing of the shot peened specimen, was attributed to increased volume fraction of stabilized nanostructure, stress recovery, and crystallization of the oxide film. Increase in corrosion resistance from annealing of shot peened material was related to more effective passivation. Thus, the surface of c p-Ti, modified by this novel process, possessed a unique quality of enhancing cell proliferation as well as the corrosion resistance and could be highly effective in reducing treatment time of patients adopting dental and orthopedic implants of titanium and its alloys.


Asunto(s)
Materiales Dentales/química , Nanoestructuras/química , Osteoblastos/fisiología , Titanio/química , Ultrasonido/métodos , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular , Colorantes , Corrosión , Cristalización , Pulido Dental/métodos , Calor , Humanos , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Nitritos/análisis , Óxidos/química , Potenciometría , Acero Inoxidable/química , Estrés Mecánico , Propiedades de Superficie , Sales de Tetrazolio , Tiazoles , Factores de Tiempo
11.
J Immunol ; 186(4): 2262-72, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21242512

RESUMEN

IL-3, a cytokine secreted by Th cells, functions as a link between the immune and the hematopoietic system. We previously demonstrated the potent inhibitory role of IL-3 on osteoclastogenesis, pathological bone resorption, and inflammatory arthritis. In this study, we investigated the novel role of IL-3 in development of regulatory T (Treg) cells. We found that IL-3 in a dose-dependent manner increases the percentage of Foxp3(+) Treg cells indirectly through secretion of IL-2 by non-Treg cells. These IL-3-expanded Treg cells are competent in suppressing effector T cell proliferation. Interestingly, IL-3 treatment significantly reduces the severity of arthritis and restores the loss of Foxp3(+) Treg cells in thymus, lymph nodes, and spleen in collagen-induced arthritis mice. Most significantly, we show that IL-3 decreases the production of proinflammatory cytokines IL-6, IL-17A, TNF-α, and IL-1 and increases the production of anti-inflammatory cytokines IFN-γ and IL-10 in collagen-induced arthritis mice. Thus, to our knowledge, we provide the first evidence that IL-3 play an important role in modulation of Treg cell development in both in vitro and in vivo conditions, and we suggest its therapeutic potential in the treatment of rheumatoid arthritis and other autoimmune diseases.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/terapia , Diferenciación Celular/inmunología , Colágeno/administración & dosificación , Factores de Transcripción Forkhead/biosíntesis , Interleucina-3/uso terapéutico , Linfocitos T Reguladores/inmunología , Animales , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Artritis Reumatoide/terapia , Interleucina-3/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Linfocitos T Reguladores/patología
12.
Biochem Biophys Res Commun ; 418(4): 669-75, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22293197

RESUMEN

IL-3 is an important cytokine that regulates hematopoiesis. We have previously demonstrated that IL-3 is a potent inhibitor of osteoclastogenesis and bone resorption. In the present study, we have investigated the role of IL-3 on human osteoblast differentiation and bone formation. We found that IL-3 in a dose-dependent manner increases osteoblast differentiation and matrix mineralization in human mesenchymal stem cells (MSCs). IL-3 significantly enhances the expression of osteoblast specific genes such as alkaline phosphatase, collagen type-I, osteocalcin and osteopontin; and Runx-2 and osterix transcription factors. Moreover, IL-3 induces the expression of bone morphogenetic protein-2 (BMP-2), and activates smad1/5/8. IL-3 enhances osteoblast differentiation and BMP-2 secretion through JAK/STAT pathway. Interestingly, IL-3 promotes in vivo bone regeneration ability of MSCs. Thus, we reveal for the first time that IL-3 enhances human osteoblast differentiation and bone formation in both in vitro and in vivo conditions, and suggest its therapeutic potential for bone formation in important bone diseases.


Asunto(s)
Diferenciación Celular , Interleucina-3/fisiología , Células Madre Mesenquimatosas/citología , Osteoblastos/citología , Osteogénesis , Fosfatasa Alcalina/biosíntesis , Animales , Proteína Morfogenética Ósea 2/biosíntesis , Regeneración Ósea , Colágeno Tipo I/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Humanos , Interleucina-3/farmacología , Subunidad alfa del Receptor de Interleucina-3/biosíntesis , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Osteocalcina/biosíntesis , Osteopontina/biosíntesis , Factor de Transcripción Sp7 , Factores de Transcripción/biosíntesis
13.
J Immunol ; 185(4): 2261-72, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20644169

RESUMEN

IL-3 is an important cytokine that regulates hematopoiesis and functions as a link between the immune and the hematopoietic system. In this study, we investigated the role and mechanism of IL-3 action on human osteoclast formation and bone resorption using PBMCs. PBMCs differentiate into functional osteoclasts in the presence of M-CSF and receptor activator of NF-kappaB ligand as evaluated by 23c6 expression and bone resorption. We found that IL-3 dose-dependently inhibited formation of 23c6-positive osteoclasts, bone resorption and C-terminal telopeptide of type I collagen, a collagen degradation product. The inhibitory effect of IL-3 on bone resorption was irreversible. To investigate the mechanism of IL-3 action, we analyzed the effect of IL-3 on the receptor activator of NF-kappaB and c-Fms receptors and c-Fos, PU.1, NFAT cytoplasmic 1, and RelB transcription factors essential for osteoclastogenesis. IL-3 significantly inhibited c-Fms and downregulated both PU.1 and c-Fos at both mRNA and protein level. Furthermore, IL-3-treated cells showed increased expression of dendritic cell markers CD1a and CD80 and decreased expression of monocyte/macrophage marker CD14. Interestingly, IL-3 inhibited formation of human osteoclasts derived from blood monocytes and bone marrow cells of osteoporotic individuals. Thus, IL-3 may have therapeutic potential as an antiosteolytic agent in treatment of osteoporosis.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Interleucina-3/farmacología , Osteoclastos/efectos de los fármacos , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Adulto , Anciano , Resorción Ósea/prevención & control , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Persona de Mediana Edad , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptores de Interleucina-3/genética , Receptores de Interleucina-3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
14.
Cancer Res ; 82(23): 4303-4312, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36191083

RESUMEN

Obesity induces multifactorial effects such as dyslipidemia, insulin resistance, and arterial hypertension that influence the progression of many diseases. Obesity is associated with an increased incidence of cancers, and multiple mechanisms link obesity with cancer initiation and progression. Macrophages participate in the homeostasis of adipose tissue and play an important role in cancer. Adipose tissue expansion in obesity alters the balance between pro- and anti-inflammatory macrophages, which is a primary cause of inflammation. Chronic low-grade inflammation driven by macrophages is also an important characteristic of cancer. Adipocytes secrete various adipokines, including adiponectin, leptin, IL6, and TNFα, that influence macrophage behavior and tumor progression. Furthermore, other metabolic effects of obesity, such as hyperlipidemia, hyperglycemia, and hypercholesterolemia, can also regulate macrophage functionality in cancer. This review summarizes how obesity influences macrophage-tumor cell interactions and the role of macrophages in the response to anticancer therapies under obese conditions.


Asunto(s)
Resistencia a la Insulina , Neoplasias , Humanos , Obesidad/patología , Macrófagos/metabolismo , Tejido Adiposo/metabolismo , Inflamación/patología , Neoplasias/patología
15.
J Immunol ; 182(1): 361-70, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109167

RESUMEN

IL-3, a cytokine secreted by activated T cells is well known to regulate the proliferation, differentiation, and survival of pluripotent hematopoietic stem cells. IL-3 functions as a link between the immune and the hematopoietic system. In this study, we suggest an important new role of IL-3 in inhibition of TNF-alpha-induced bone resorption in vitro and prevention of inflammatory arthritis in mice. We show here that IL-3 potently and irreversibly inhibits TNF-alpha-induced bone resorption in hematopoietic precursors of monocyte/macrophage lineage. IL-3 showed an inhibitory effect on TNF-alpha-induced bone resorption even in the presence of proinflammatory cytokines such as IL-1alpha, TGF-beta(1), TGF-beta(3), IL-6, and PGE(2). We found that IL-3 prevented TNF-alpha-induced c-fos nuclear translocation and AP-1 DNA-binding activity. Interestingly, IL-3 pretreatment prevented the development of inflammatory arthritis in mice induced by a mixture of anti-type II collagen mAbs and LPS. Furthermore, IL-3 prevented cartilage and bone loss in the joints indirectly through inhibition of inflammation. Thus, we provide the first evidence that IL-3, a strong regulator of hematopoiesis, also plays an important role in inhibition of TNF-alpha-induced bone resorption and prevention of inflammatory arthritis in mice.


Asunto(s)
Artritis Experimental/inmunología , Artritis Experimental/prevención & control , Resorción Ósea/inmunología , Resorción Ósea/prevención & control , Mediadores de Inflamación/fisiología , Interleucina-3/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Transporte Activo de Núcleo Celular/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Artritis Experimental/metabolismo , Resorción Ósea/patología , Cartílago Articular/inmunología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Células Cultivadas , Colágeno Tipo II/inmunología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Mediadores de Inflamación/administración & dosificación , Interleucina-3/administración & dosificación , Lipopolisacáridos/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Técnicas de Cultivo de Órganos , Osteocondritis/inmunología , Osteocondritis/metabolismo , Osteocondritis/prevención & control , Unión Proteica/genética , Unión Proteica/inmunología , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
J Biosci ; 462021.
Artículo en Inglés | MEDLINE | ID: mdl-34183475

RESUMEN

IL-3, a haematopoiesis regulatory factor, has previously been shown to inhibit both mouse and human osteoclast differentiation and bone resorption. Here, the role of rat IL-3 on rat osteoclast differentiation was evaluated to address whether the inhibitory action of IL-3 on osteoclastogenesis is conserved in various species. It was observed that IL-3 inhibited rat osteoclast differentiation induced by both TNF-α and receptor activator of NF-ĸB ligand (RANKL). TNF-α is known to induce bone loss in postmenopausal osteoporotic women and it also synergise with many pro-osteoclastogenic cytokines to cause huge pathological bone loss. Importantly, it was found that rat IL-3 inhibits the synergistic action of TNF-α with RANKL and IL-1ß, TGFß1 and TGF-ß3. IL-3 downregulates the TNF-α-induced nuclear translocation of NF-ĸB-p65 and c-fos without affecting c-jun. Interestingly, we observed that IL-3 also inhibits osteoclast differentiation in vivo in rats induced by TNF-α. All these results suggest that inhibitory action of IL-3 on osteoclastogenesis is conserved in various species including mice, rats and humans. Thus, our results clearly indicate that IL-3 has therapeutic potential to treat pathological bone loss in important skeletal diseases.


Asunto(s)
Diferenciación Celular , Interleucina-3/fisiología , Osteoclastos , Osteogénesis , Animales , FN-kappa B/metabolismo , Ligando RANK/metabolismo , Ratas Wistar , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Biochem Biophys Res Commun ; 399(4): 688-93, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20691668

RESUMEN

IL-3, a cytokine secreted by activated T lymphocytes, stimulates the proliferation, differentiation and survival of pluripotent hematopoietic stem cells. In this study, we investigated the mechanism of inhibitory action of IL-3 on osteoclast differentiation. We show here that IL-3 significantly inhibits receptor activator of NF-kappaB (RANK) ligand (RANKL)-induced activation of c-Jun N-terminal kinase (JNK). IL-3 down-regulates expression of c-Fos and nuclear factor of activated T cells (NFATc1) transcription factors. In addition, IL-3 down-regulates RANK expression posttranscriptionally in both purified osteoclast precursors and whole bone marrow cells. Furthermore, the inhibitory effect of IL-3 on RANK expression was irreversible. Interestingly, IL-3 inhibits in vivo RANK expression in mice. Thus, we provide the first evidence that IL-3 irreversibly inhibits RANK expression that results in inhibition of important signaling molecules induced by RANKL.


Asunto(s)
Diferenciación Celular , Interleucina-3/metabolismo , Osteoclastos/citología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/biosíntesis , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Interleucina-3/farmacología , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/biosíntesis , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores
18.
Biochem Biophys Res Commun ; 393(3): 377-83, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20138833

RESUMEN

Mesenchymal stem cells (MSCs) are capable of self-renewal and differentiation into multiple cell lineages. Presently, bone marrow is considered as a prime source of MSCs; however, there are some drawbacks and limitations in use of these MSCs for cell therapy. In this study, we demonstrate that human gingival tissue-derived MSCs have several advantages over bone marrow-derived MSCs. Gingival MSCs are easy to isolate, homogenous and proliferate faster than bone marrow MSCs without any growth factor. Importantly, gingival MSCs display stable morphology and do not loose MSC characteristic at higher passages. In addition, gingival MSCs maintain normal karyotype and telomerase activity in long-term cultures, and are not tumorigenic. Thus, we reveal that human gingiva is a better source of MSCs than bone marrow, and large number of functionally competent clinical grade MSCs can be generated in short duration for cell therapy in regenerative medicine and tissue engineering.


Asunto(s)
Separación Celular/métodos , Encía/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Células de la Médula Ósea/fisiología , Regeneración Ósea , Diferenciación Celular , Transformación Celular Neoplásica , Humanos , Cariotipificación , Medicina Regenerativa
19.
Ultrasonics ; 104: 106110, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32146383

RESUMEN

Surface mechanical attrition treatment (SMAT) of metallic biomaterials has gained significant importance due to its ability to develop nano structure in the surface region. In the present study, the microstructural changes and corrosion behavior of the commercially pure titanium (cp-Ti), following different durations of ultrasonic shot peening (USSP) has been investigated. cp-Ti was shot peened for different durations from 0 to 120 s and the treated samples were examined for microstructural changes in the surface region, cell viability and corrosion behavior. Cell viability was considerably increased after USSP for 60-120 s, exhibiting maximum for the 90 s of USSP. The passivation tendency was also improved with peening duration up to 90 s, however, it declined for longer duration of USSP. The beneficial effects of USSP may be attributed to nano structuring in the surface region and development of higher positive potentials at the USSP treated surface. Transmission Electron Microscope (TEM) examination of the USSPed surface revealed dislocation entanglement and substructure. Also, higher surface volta potential was observed over the USSPed sample exhibiting better cell proliferation. The present work is corollary to previous work of the group and mainly discusses the role of USSP duration, as a process parameter, on the cell viability and corrosion resistance of cp-Ti.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Nanoestructuras/química , Titanio/química , Ultrasonido/métodos , Proliferación Celular , Supervivencia Celular , Corrosión , Técnicas Electroquímicas , Humanos , Ensayo de Materiales , Microscopía de Fuerza Atómica , Microscopía Electrónica , Propiedades de Superficie , Difracción de Rayos X
20.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 547-557, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30579930

RESUMEN

Estrogen insufficiency at menopause cause accelerated bone loss due to unwarranted differentiation and function of osteoclasts. Unraveling the underlying mechanism/s may identify mediators of estrogen action which can be targeted for improved management of osteoporosis. Towards this, we analyzed the effect of 17ß-estradiol on the proteomes of differentiating human osteoclasts. The major proteomic changes observed included upregulation of LYN by estrogen. We, therefore, investigated the effect of estrogen on osteoclast differentiation, survival, and function in control and LYN knockdown conditions. In control condition, estrogen treatment increased the apoptosis rate and suppressed the calcium signaling by reducing the intracellular Ca2+ levels as well as expression and activation of NFATc1 and c-Src during differentiation, resulting in reduced osteoclastogenesis. These osteoclasts were smaller in size with reduced extent of multinuclearity and produced significantly low levels of bone resorbing enzymes. They also exhibited disrupted sealing zone formation with low podosome density, impaired cell polarization and reduced resorption of dentine slices. Interestingly, in LYN knockdown condition, estrogen failed to induce apoptosis and inhibit activation of NFATc1 and c-Src. Compared to effect of estrogen on osteoclast in control condition, LYN knockdown osteoclasts did not show reduction in production of bone resorbing enzymes and had defined sealing zone formation with high podosome density with no impairment in cell polarization. They resorbed significant area on dentine slices. Thus, the inhibitory action of estrogen on osteoclast was severely restrained in LYN knockdown condition, demonstrating the importance of LYN as a key mediator of the effect of estrogen on osteoclastogenesis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Estradiol/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Familia-src Quinasas/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Desarrollo Óseo/efectos de los fármacos , Desarrollo Óseo/genética , Resorción Ósea/genética , Resorción Ósea/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Osteoclastos/metabolismo , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA