Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Mol Biol Rep ; 47(12): 9659-9665, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33269432

RESUMEN

Multiple sclerosis (MS) is an autoimmune-type inflammatory disorder in human central nervous system. Recombinant interferon beta (IFN-ß) decreases the number of relapses and postpones disability progression in MS. However, up to 50% of patients treated with interferon beta continue experiencing relapses and/or worsening disability. Single nucleotide polymorphisms in different genes have been known to show significant associations with response to IFN-ß in MS patients. In the present work, we examined the potential role of TRAILR1 and GRIA3 genes polymorphisms on response to IFN-ß therapy in Iranian MS patients. The DNA was extracted from blood samples by standard procedures from 73 patients diagnosed with Multiple Sclerosis that were either responded to IFN-ß or did not. We carried out RFLP -PCR and tetra-primer ARMS-PCR methods to study of rs20576 and rs12557782, respectively. All results were analyzed using the SPSS software. TRAILR1 rs20576 genotype frequencies in responders and non-responders were similar (χ2 = 0.26, P = 0.87, Fisher, s Exact test). Our results showed that response to IFN-ß has not association with sex (p = 0.73). Also, genotypic frequencies of GRIA3 rs12557782 had no significant differences between two groups of female population (χ2 = 3.75, p = 0.15). Furthermore, it had not been any statistical differences between responder and non-responder males (χ2 = 0.7, p = 0.4) related to the SNP. Our results analysis revealed no significant association between the studied SNPs (TRAILR1 rs20576 and GRIA3rs 12,557,782) and response to IFN-ß in Iranian MS patients.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Interferón beta/uso terapéutico , Esclerosis Múltiple Recurrente-Remitente/genética , Polimorfismo de Nucleótido Simple , Receptor Activador del Factor Nuclear kappa-B/genética , Receptores AMPA/genética , Adolescente , Adulto , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Masculino , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Farmacogenética , Polimorfismo de Longitud del Fragmento de Restricción , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptores AMPA/inmunología , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
2.
Eur J Pharmacol ; 879: 173129, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32339516

RESUMEN

The main aims of this study were: (1) to investigate whether a blockade of the interaction between the receptor activator of nuclear factor-κB (NF-ĸB) ligand (RANKL) and its receptor RANK may have potential as a novel therapeutic strategy for allergic asthma; (2) to compare the efficacies of the blockade of RANKL/RANK interaction as well as the blockade of NF-κB inhibitor kinase (IKK) and of NF-κB translocation to the nucleus, also in comparison with glucocorticosteroid treatment, in terms of the development of a mouse model of allergic airway inflammation (AAI) and accompanying immune response. The blockade of each of the targets fully prevented the development of AAI. All the tested therapeutic strategies seemed to have a certain advantage over glucocorticosteroids with regard to counteracting the development of AAI. Prevention of the activation and clonal expansion of CD4+ effector T (Teff) cells in the mediastinal lymph nodes (MLNs) constitutes a fundamental event underlying the anti-asthmatic effect induced by the blockade of IKK, NF-κB translocation or of RANKL/RANK interaction. The results indicate that attenuation of the CD11b+CD103-CD11chigh dendritic cell response in the MLNs is an initial but not the main mechanism responsible for this effect. In turn, the direct anti-proliferative action on CD4+ Teff cells seems to constitute the chief mechanism responsible for the anti-asthmatic effect of all the tested therapeutic strategies. A clinical implication is that local inhibition of RANKL/RANK interaction achieved via inhalatory administration of a RANKL antagonist can be considered as a novel therapeutic strategy in treatment of allergic asthma.


Asunto(s)
Asma/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Ligando RANK/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Animales , Asma/inmunología , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones Endogámicos BALB C , FN-kappa B/inmunología , Ovalbúmina , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
3.
Front Immunol ; 11: 623265, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33552088

RESUMEN

Thymic epithelial cells (TECs) provide essential clues for the proliferation, survival, migration, and differentiation of thymocytes. Recent advances in mouse and human have revealed that TECs constitute a highly heterogeneous cell population with distinct functional properties. Importantly, TECs are sensitive to thymic damages engendered by myeloablative conditioning regimen used for bone marrow transplantation. These detrimental effects on TECs delay de novo T-cell production, which can increase the risk of morbidity and mortality in many patients. Alike that TECs guide the development of thymocytes, reciprocally thymocytes control the differentiation and organization of TECs. These bidirectional interactions are referred to as thymic crosstalk. The tumor necrosis factor receptor superfamily (TNFRSF) member, receptor activator of nuclear factor kappa-B (RANK) and its cognate ligand RANKL have emerged as key players of the crosstalk between TECs and thymocytes. RANKL, mainly provided by positively selected CD4+ thymocytes and a subset of group 3 innate lymphoid cells, controls mTEC proliferation/differentiation and TEC regeneration. In this review, I discuss recent advances that have unraveled the high heterogeneity of TECs and the implication of the RANK-RANKL signaling axis in TEC differentiation and regeneration. Targeting this cell-signaling pathway opens novel therapeutic perspectives to recover TEC function and T-cell production.


Asunto(s)
Células Epiteliales/fisiología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Regeneración/inmunología , Transducción de Señal , Timo/fisiología , Humanos , Ligando RANK/inmunología , Timocitos/inmunología
4.
Front Immunol ; 10: 1323, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244859

RESUMEN

Microfold (M) cells residing in the follicle-associated epithelium of mucosa-associated lymphoid tissues are specialized for sampling luminal antigens to initiate mucosal immune responses. In the past decade, glycoprotein 2 (GP2) and Tnfaip2 were identified as reliable markers for M cells in the Peyer's patches of the intestine. Furthermore, RANKL-RANK signaling, as well as the canonical and non-canonical NFκB pathways downstream, is essential for M-cell differentiation from the intestinal stem cells. However, the molecular characterization and differentiation mechanisms of M cells in the lower respiratory tract, where organized lymphoid tissues exist rarely, remain to be fully elucidated. Therefore, this study aimed to explore M cells in the lower respiratory tract in terms of their specific molecular markers, differentiation mechanism, and functions. Immunofluorescence analysis revealed a small number of M cells expressing GP2, Tnfaip2, and RANK is present in the lower respiratory tract of healthy mice. The intraperitoneal administration of RANKL in mice effectively induced M cells, which have a high capacity to take up luminal substrates, in the lower respiratory epithelium. The airway M cells associated with lymphoid follicles were frequently detected in the pathologically induced bronchus-associated lymphoid tissue (iBALT) in the murine models of autoimmune disease as well as pulmonary emphysema. These findings demonstrate that RANKL is a common inducer of M cells in the airway and digestive tracts and that M cells are associated with the respiratory disease. We also established a two-dimensional culture method for airway M cells from the tracheal epithelium in the presence of RANKL successfully. This model may be useful for functional studies of M cells in the sampling of antigens at airway mucosal surfaces.


Asunto(s)
Inmunidad Mucosa , Ligando RANK/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Enfermedades Respiratorias/inmunología , Enfermedades Respiratorias/patología , Animales , Bronquiolos/inmunología , Bronquiolos/patología , Técnicas de Cultivo de Célula , Fumar Cigarrillos/efectos adversos , Fumar Cigarrillos/inmunología , Fumar Cigarrillos/patología , Modelos Animales de Enfermedad , Enfisema/inmunología , Enfisema/patología , Femenino , Proteínas Ligadas a GPI/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal/inmunología , Factores de Necrosis Tumoral/inmunología
5.
Front Immunol ; 10: 951, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31114586

RESUMEN

The maintenance of B cell homeostasis requires a tight control of B cell generation, survival, activation, and maturation. In lymphocytes upon activation, increased sensitivity to apoptotic signals helps controlling differentiation and proliferation. The death receptor Fas is important in this context because genetic Fas mutations in humans lead to an autoimmune lymphoproliferative syndrome that is similar to lymphoproliferation observed in Fas-deficient mice. In contrast, the physiological role of TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs) in humans has been poorly studied so far. Indeed, most studies have focused on tumor cell lines and on mouse models whose results are difficult to transpose to primary human B cells. In the present work, the expression of apoptosis-inducing TRAIL-R1 and TRAIL-R2 and of the decoy receptors TRAIL-R3 and TRAIL-R4 was systematically studied in all developmental stages of peripheral B cells isolated from the blood and secondary lymphoid organs. Expression of TRAIL-Rs is modulated along development, with highest levels observed in germinal center B cells. In addition, T-dependent and T-independent signals elicited induction of TRAIL-Rs with distinct kinetics, which differed among B cell subpopulations: switched memory cells rapidly upregulated TRAIL-R1 and -2 upon activation while naïve B cells only reached similar expression levels at later time points in culture. Increased expression of TRAIL-R1 and -2 coincided with a caspase-3-dependent sensitivity to TRAIL-induced apoptosis in activated B cells but not in freshly isolated resting B cells. Finally, both TRAIL-R1 and TRAIL-R2 could signal actively and both contributed to TRAIL-induced apoptosis. In conclusion, this study provides a systematic analysis of the expression of TRAIL-Rs in human primary B cells and of their capacity to signal and induce apoptosis. This dataset forms a basis to further study and understand the dysregulation of TRAIL-Rs and TRAIL expression observed in autoimmune diseases. Additionally, it will be important to foresee potential bystander immunomodulation when TRAIL-R agonists are used in cancer treatment.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Activación de Linfocitos , Proteínas de la Membrana/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Linfocitos B/citología , Caspasa 3/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Masculino , Transducción de Señal/inmunología
6.
Int J Mol Sci ; 20(9)2019 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-31052546

RESUMEN

RANK ligand (RANKL) is a member of the tumor necrosis factor alpha superfamily of cytokines. It is the only known ligand binding to a membrane receptor named receptor activator of nuclear factor-kappa B (RANK), thereby triggering recruitment of tumor necrosis factor (TNF) receptor associated factor (TRAF) adaptor proteins and activation of downstream pathways. RANK/RANKL signaling is controlled by a decoy receptor called osteoprotegerin (OPG), but also has additional more complex levels of regulation. The existing literature on RANK/RANKL signaling in cervical cancer was reviewed, particularly focusing on the effects on the microenvironment. RANKL and RANK are frequently co-expressed in cervical cancer cells lines and in carcinoma of the uterine cervix. RANKL and OPG expression strongly increases during cervical cancer progression. RANKL is directly secreted by cervical cancer cells, which may be a mechanism they use to create an immune suppressive environment. RANKL induces expression of multiple activating cytokines by dendritic cells. High RANK mRNA levels and high immunohistochemical OPG expression are significantly correlated with high clinical stage, tumor grade, presence of lymph node metastases, and poor overall survival. Inhibition of RANKL signaling has a direct effect on tumor cell proliferation and behavior, but also alters the microenvironment. Abundant circumstantial evidence suggests that RANKL inhibition may (partially) reverse an immunosuppressive status. The use of denosumab, a monoclonal antibody directed to RANKL, as an immunomodulatory strategy is an attractive concept which should be further explored in combination with immune therapy in patients with cervical cancer.


Asunto(s)
Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Neoplasias del Cuello Uterino/inmunología , Animales , Cuello del Útero/inmunología , Cuello del Útero/patología , Femenino , Humanos , Inmunoterapia/métodos , Ligando RANK/análisis , Receptor Activador del Factor Nuclear kappa-B/análisis , Transducción de Señal , Microambiente Tumoral , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia
7.
Front Immunol ; 10: 629, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30984193

RESUMEN

The identification of Receptor activator of nuclear factor kappa B ligand (RANKL) and its cognate receptor Receptor activator of nuclear factor kappa B (RANK) during a search for novel tumor necrosis factor receptor (TNFR) superfamily members has dramatically changed the scenario of bone biology by providing the functional and biochemical proof that RANKL signaling via RANK is the master factor for osteoclastogenesis. In parallel, two independent studies reported the identification of mouse RANKL on activated T cells and of a ligand for osteoprotegerin on a murine bone marrow-derived stromal cell line. After these seminal findings, accumulating data indicated RANKL and RANK not only as essential players for the development and activation of osteoclasts, but also for the correct differentiation of medullary thymic epithelial cells (mTECs) that act as mediators of the central tolerance process by which self-reactive T cells are eliminated while regulatory T cells are generated. In light of the RANKL-RANK multi-task function, an antibody targeting this pathway, denosumab, is now commonly used in the therapy of bone loss diseases including chronic inflammatory bone disorders and osteolytic bone metastases; furthermore, preclinical data support the therapeutic application of denosumab in the framework of a broader spectrum of tumors. Here, we discuss advances in cellular and molecular mechanisms elicited by RANKL-RANK pathway in the bone and thymus, and the extent to which its inhibition or augmentation can be translated in the clinical arena.


Asunto(s)
Enfermedades Óseas/inmunología , Huesos/inmunología , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal/inmunología , Timo/inmunología , Animales , Enfermedades Óseas/patología , Enfermedades Óseas/terapia , Huesos/patología , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Osteoclastos/inmunología , Osteoclastos/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Timo/patología
8.
Crit Rev Oncol Hematol ; 133: 85-91, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30661662

RESUMEN

Binding between the receptor activator of nuclear factor-kB (RANK) and its ligand (RANKL) triggers recruitment of TNF receptor associated factor (TRAF) adaptor proteins and activation of downstream pathways. RANK/RANKL signaling is controlled by a decoy receptor called osteoprotegerin (OPG) which interacts with RANKL. Additional networks regulating RANK/RANKL signaling are active in a context specific manner. RANK/RANKL signaling is essential for the differentiation of bone-resorbing osteoclasts, and is deregulated in pathological processes such as postmenopausal osteoporosis or cancer induced bone destruction. Cells expressing RANK and RANKL are commonly found in the tumor microenvironment. The RANKL/RANK pathway is often overexpressed in tumors of the breast, prostate, endometrium, cervix, stomach, oesophagus and bladder, thyroid and correlated with poor prognosis. RANK signaling plays an important role in the innate and adaptive immune response as it generates regulatory T (Treg) cells and increases production of cytokines. RANK expression induces chemoresistance in vitro through the activation of multiple signal transduction pathways. RANKL blockade improves the efficacy of anti-CTLA-4 monoclonal antibodies against solid tumors and experimental metastases. As RANK inhibition enhances the immune response there is an increasing interest in combining it with immune therapy in an attempt to sensitize immune resistant tumors to immune therapies. Several studies are ongoing to assess this concept. The role of RANK/RANKL inhibition should be further pursued as an immunomodulatory strategy in combination with other treatment modalities.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Denosumab/administración & dosificación , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Ligando RANK/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Denosumab/farmacología , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Ligando RANK/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
9.
Nat Rev Clin Oncol ; 15(11): 676-693, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30232468

RESUMEN

Recognizing that the transformative effects of immunotherapy are currently limited to a minority of patients with cancer, research efforts are increasingly focused on expanding and enhancing clinical responses by combining immunotherapies; the repurposing of existing drugs is an attractive approach, given their well-characterized safety and pharmacokinetic profiles. Receptor activator of nuclear factor-κB (RANK) and the RANK ligand (RANKL) were initially described in the context of T cell-dendritic cell interactions; however, the discovery of an obligate role of RANK signalling in osteoclastogenesis led to the development of the anti-RANKL antibody denosumab for antiresorptive indications, including bone metastases. Randomized clinical trials and post-marketing surveillance studies have established the acceptable safety profile of denosumab. More recently, several case reports involving patients with advanced-stage melanoma have described remarkable responses following concurrent treatment with denosumab and immune-checkpoint inhibitors. Randomized trials assessing similar combinations in patients with melanoma or renal cell carcinoma are now underway. Herein, we discuss the hallmark clinical trials of denosumab in light of possible immunological effects of this agent. We highlight the role of immune cells as sources of RANK and RANKL in the tumour microenvironment and review data on RANKL inhibition in mouse models of cancer. Finally, we describe hypothetical immune-related mechanisms of action, which could be assessed in clinical trials of immune-checkpoint inhibitors and denosumab in patients with cancer.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Carcinoma de Células Renales/inmunología , Denosumab/uso terapéutico , Humanos , Melanoma/inmunología , Osteogénesis/inmunología , Ligando RANK/antagonistas & inhibidores , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Microambiente Tumoral/inmunología
10.
Int J Mol Med ; 42(4): 2071-2079, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30066839

RESUMEN

Osteoimmunological studies have revealed that T cells exert a powerful impact on the formation and activity of osteoclasts and bone remodeling. Evidence demonstrates that immature dendritic cells (iDCs) are more efficient transdifferentiating into osteoclasts (OCs) than monocytes. However, whether Vγ9Vδ2 T (γδ T) cells stimulate or inhibit iDC transdifferentiation into OCs has never been reported. The aim of the present study was to investigate the effects of γδ T cells on this transdifferentiation process. γδ T cells and iDCs were isolated from the peripheral blood of healthy volunteers separately and were co­cultured with Transwelll inserts, with γδ T cells in the upper chamber and iDCs in the lower chamber. IDCs were treated with macrophage­colony stimulating factor and receptor activator of nuclear factor­κB (RANK) ligand. Tartrate resistant acid phosphatase (TRAP) assay and dentine resorption assay were performed to detect OC formation and their resorption capacity, respectively. The mRNA expression of OCs was examined using a microarray and real time­quantitative polymerase chain reaction to trace the changes during iDC transdifferentiation into OCs. The results demonstrated that γδ T cells significantly inhibited the generation of the TRAP­positive OCs from iDCs and their resorption capacity. The microarray analysis identified decreased expression level of Fos proto­oncogene AP­1 transcription factor subunit (c­Fos), ATPase H+ transporting V0 subunit d (ATP6V0D2) and cathepsin K when iDCs were co­cultured with γδ T cells. These genes are associated with OC differentiation, indicating that γδ T cells suppressed iDCs osteoclastogenesis by downregulation of the RANK/c­Fos/ATP6V0D2 signaling pathway. The present findings provide novel insights into the interactions between human γδ T cells and iDCs, and demonstrate that γδ T cells are capable of inhibiting OC formation and their activity via downregulation of genes associated with OC differentiation.


Asunto(s)
Transdiferenciación Celular/inmunología , Células Dendríticas/inmunología , Osteoclastos/inmunología , Proteínas Proto-Oncogénicas c-fos/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , ATPasas de Translocación de Protón Vacuolares/inmunología , Células Dendríticas/patología , Humanos , Osteoclastos/patología , Linfocitos T/patología
11.
Neuroimmunomodulation ; 25(1): 23-33, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29920500

RESUMEN

OBJECTIVES: Our study focused on the RANKL (receptor activator of nuclear factor-κB ligand)/RANK/OPG (osteoprotegerin) axis and selected proinflammatory/immunoregulatory upstream mediators in the peripheral blood (PBL) and cerebrospinal fluid (CSF) of multiple sclerosis (MS) patients. METHODS: PBL and CSF were collected from healthy controls (n = 35) and MS patients at the clinical onset of the disease (n = 33). In addition, PBL samples were obtained from relapse-remitting (RR)-MS patients (n = 30). Patients were assessed by means of the expanded disability status scale (EDSS) and routine laboratory parameters. Soluble (s)RANKL and OPG were measured in the CSF and plasma; gene expression was detected for RANKL, RANK, OPG, and selected cytokines/chemokines (interleukin [IL]-4, IL-10, IL-17, CCL2, and CXCL12) in PBL mononuclear cells. RESULTS: The OPG level in the CSF was lower in MS patients at clinical onset than in controls. Moreover, the sRANKL/OPG ratio was higher in the CSF of MS patients at clinical onset and in the plasma of RR-MS patients than in controls. Gene expression of RANKL/RANK/OPG in PBL mononuclear cells was higher only in RR-MS patients. IL-4, CCL2, and CXCL12 were positively correlated and IL-10 was negatively correlated with RANKL/RANK expression. OPG was negatively correlated with EDSS and alkaline phosphatase level. CONCLUSION: Our study revealed that changes of RANKL/RANK/OPG axis are associated with MS, particularly the decreased OPG level in the CSF at disease onset. Therefore, these factors may serve as disease biomarkers and molecular targets of novel therapeutic approaches.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente/líquido cefalorraquídeo , Esclerosis Múltiple Recurrente-Remitente/diagnóstico , Osteoprotegerina/líquido cefalorraquídeo , Ligando RANK/líquido cefalorraquídeo , Receptor Activador del Factor Nuclear kappa-B/líquido cefalorraquídeo , Adulto , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/inmunología , Osteoprotegerina/inmunología , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología
12.
Food Funct ; 9(5): 2653-2660, 2018 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-29666861

RESUMEN

Osteoporosis can be classified as an inflammatory disease and the crosstalk between the immune system and bone growth should be considered. The effects of bovine lactoferrin on bone by osteoimmunology were investigated in the present study. Ten week old female BALB/c mice were ovariectomized (OVX) and fed for 12 weeks with a control diet or lactoferrin (2, 20 and 100 mg kg-1 d-1). The results showed that following 12 weeks of treatment after surgery, OVX resulted in bone loss, but treatment with lactoferrin preserved bone homeostasis. Lactoferrin significantly improved bone mineral density, and increased the serum levels of alkaline phosphatase, while it decreased the serum levels of tartrate-resistant acid phosphatase. Furthermore, according to micro-computed tomography (micro-CT), the bone volume per tissue volume (BV/TV), trabecular thickness (Tb.Th) and trabecular number (Tb.N) were elevated. The results indicated that the structure model index (SMI) was reduced in the OVX + LF groups. Additionally, lactoferrin enhanced the Max-Load and Max-Stress values of the femur, and increased the content of Ca and P. However, lactoferrin suppressed the RANKL/OPG ratio in OVX mice. Moreover, interferon-γ, interleukin-5 and interleukin-10 were elevated significantly in the OVX + LF groups. Lactoferrin had a positive effect on the bone micro-environment and might be a pleiotropic protein for the prevention and treatment of estrogen-dependent bone loss via the osteoimmunology pathway.


Asunto(s)
Lactoferrina/administración & dosificación , Osteoporosis/prevención & control , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Animales , Densidad Ósea , Huesos/efectos de los fármacos , Huesos/inmunología , Bovinos , Femenino , Homeostasis/efectos de los fármacos , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Ratones , Ratones Endogámicos BALB C , Osteoporosis/genética , Osteoporosis/inmunología , Osteoporosis/fisiopatología , Ovariectomía , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética
14.
J Immunol ; 200(1): 218-228, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29141864

RESUMEN

As osteoclasts have the central roles in normal bone remodeling, it is ideal to regulate only the osteoclasts performing pathological bone destruction without affecting normal osteoclasts. Based on a hypothesis that pathological osteoclasts form under the pathological microenvironment of the bone tissues, we here set up optimum culture conditions to examine the entity of pathologically activated osteoclasts (PAOCs). Through searching various inflammatory cytokines and their combinations, we found the highest resorbing activity of osteoclasts when osteoclasts were formed in the presence of M-CSF, receptor activator of NF-κB ligand, and IL-1ß. We have postulated that these osteoclasts are PAOCs. Analysis using confocal laser microscopy revealed that PAOCs showed extremely high proton secretion detected by the acid-sensitive fluorescence probe Rh-PM and bone resorption activity compared with normal osteoclasts. PAOCs showed unique morphology bearing high thickness and high motility with motile cellular processes in comparison with normal osteoclasts. We further examined the expression of Kindlin-3 and Talin-1, essential molecules for activating integrin ß-chains. Although normal osteoclasts express high levels of Kindlin-3 and Talin-1, expression of these molecules was markedly suppressed in PAOCs, suggesting the abnormality in the adhesion property. When whole membrane surface of mature osteoclasts was biotinylated and analyzed, the IL-1ß-induced cell surface protein was detected. PAOCs could form a subpopulation of osteoclasts possibly different from normal osteoclasts. PAOC-specific molecules could be an ideal target for regulating pathological bone destruction.


Asunto(s)
Resorción Ósea/inmunología , Interleucina-1beta/inmunología , Osteoclastos/inmunología , Animales , Adhesión Celular , Células Cultivadas , Regulación hacia Abajo , Factor Estimulante de Colonias de Macrófagos/inmunología , Masculino , Ratones , Ratones Mutantes , Terapia Molecular Dirigida , Receptor Activador del Factor Nuclear kappa-B/inmunología , Talina/genética , Talina/metabolismo
15.
Biochem Biophys Res Commun ; 493(1): 437-443, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28882590

RESUMEN

The gut-associated lymphoid tissues (GALTs), including Peyer's patches (PPs), cryptopatches (CPs) and isolated lymphoid follicles (ILFs), establish a host-microbe symbiosis by the promotion of immune reactions against gut microbes. Microfold cell inducer (MCi) cells in GALTs are the recently identified mesenchymal cells that express the cytokine RANKL and initiate bacteria-specific immunoglobulin A (IgA) production via induction of microfold (M) cell differentiation. In the previous study, the Twist2-Cre driver was utilized for gene deletion in mesenchymal cells including MCi cells. In order to investigate MCi cells more extensively, it will be necessary to develop experimental tools in addition to the Twist2-Cre driver mice and characterize such drivers in specificity and efficiency. Here we show that M cell differentiation and IgA production are impaired in the targeted deletion of RANKL by the Col6a1-Cre driver. We compared Col6a1-Cre with Twist2-Cre in terms of the specificity for mesenchymal cells in GALTs. Col6a1-Cre CAG-CAT-EGFP mice exhibited EGFP expression in podoplanin+CD31- cells including MCi cells, while Twist2-Cre mice were shown to target endothelial cells and podoplanin+CD31- cells. Tnfsf11fl/ΔCol6a1-Cre mice exhibited the absence of M cells and severe IgA reduction together with an alteration in gut microbial composition. Moreover, we analyzed germ free mice to test whether changes in the microbiota are the cause of M cell deficiency. M cell differentiation was normal in the CPs/ILFs of germ free mice, indicating that MCi cells induce M cells independently of microbial colonization. This study demonstrates that Col6a1-Cre driver mice are as useful as Twist2-Cre driver mice for functional analyses of GALT-resident mesenchymal cells, including MCi cells.


Asunto(s)
Colágeno Tipo VI/genética , Integrasas/genética , Mucosa Intestinal/inmunología , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Células Cultivadas , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL
16.
Physiol Rev ; 97(4): 1295-1349, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28814613

RESUMEN

The immune and skeletal systems share a variety of molecules, including cytokines, chemokines, hormones, receptors, and transcription factors. Bone cells interact with immune cells under physiological and pathological conditions. Osteoimmunology was created as a new interdisciplinary field in large part to highlight the shared molecules and reciprocal interactions between the two systems in both heath and disease. Receptor activator of NF-κB ligand (RANKL) plays an essential role not only in the development of immune organs and bones, but also in autoimmune diseases affecting bone, thus effectively comprising the molecule that links the two systems. Here we review the function, gene regulation, and signal transduction of osteoimmune molecules, including RANKL, in the context of osteoclastogenesis as well as multiple other regulatory functions. Osteoimmunology has become indispensable for understanding the pathogenesis of a number of diseases such as rheumatoid arthritis (RA). We review the various osteoimmune pathologies, including the bone destruction in RA, in which pathogenic helper T cell subsets [such as IL-17-expressing helper T (Th17) cells] induce bone erosion through aberrant RANKL expression. We also focus on cellular interactions and the identification of the communication factors in the bone marrow, discussing the contribution of bone cells to the maintenance and regulation of hematopoietic stem and progenitors cells. Thus the time has come for a basic reappraisal of the framework for understanding both the immune and bone systems. The concept of a unified osteoimmune system will be absolutely indispensable for basic and translational approaches to diseases related to bone and/or the immune system.


Asunto(s)
Inmunidad , Esqueleto/inmunología , Alergia e Inmunología , Animales , Artritis Reumatoide/inmunología , Comunicación Celular , Células Madre Hematopoyéticas/fisiología , Humanos , Osteoclastos/metabolismo , Osteología , Osteoprotegerina/metabolismo , Ligando RANK/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal , Esqueleto/metabolismo
17.
Clin Cancer Res ; 23(19): 5789-5801, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28634284

RESUMEN

Purpose: Novel partners for established immune checkpoint inhibitors in the treatment of cancer are needed to address the problems of primary and acquired resistance. The efficacy of combination RANKL and CTLA4 blockade in antitumor immunity has been suggested by recent case reports in melanoma. Here, we provide a rationale for this combination in mouse models of cancer.Experimental Design: The efficacy and mechanism of a combination of RANKL and CTLA4 blockade was examined by tumor-infiltrating lymphocyte analysis, tumor growth, and metastasis using a variety of neutralizing antibodies and gene-targeted mice.Results: RANKL blockade improved the efficacy of anti-CTLA4 mAbs against solid tumors and experimental metastases, with regulatory T-cell (Treg)-depleting anti-CTLA4 mAbs of the mouse IgG2a isotype showing greatest combinatorial activity. The optimal combination depended on the presence of activating Fc receptors and lymphocytes (NK cells for metastatic disease and predominantly CD8+ T cells for subcutaneous tumor control), whereas anti-RANKL alone did not require FcR. The significantly higher T-cell infiltration into solid tumors post anti-RANKL and anti-CTLA4 was accompanied by increased T-cell effector function (cytokine polyfunctionality), and anti-RANKL activity occurred independently of Treg depletion. The majority of RANKL expression in tumors was on T cells whereas RANK-expressing cells were mostly tumor-associated macrophages (TAM), with some expression also observed on dendritic cells (DC) and myeloid-derived suppressor cells (MDSC).Conclusions: These results provide a rationale for the further investigation of RANKL-RANK interactions in tumor immunity and a basis for development of translational markers of interest in human clinical trials. Clin Cancer Res; 23(19); 5789-801. ©2017 AACR.


Asunto(s)
Antígeno CTLA-4/inmunología , Inmunoterapia , Melanoma Experimental/terapia , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/genética , Citocinas/genética , Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/inmunología , Linfocitos T Reguladores/inmunología
18.
Mol Med Rep ; 14(4): 3620-6, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27572279

RESUMEN

The aim of the present study was to determine the effects of curcumin on the osteoclastogenic potential of peripheral blood mononuclear cells (PBMCs) obtained from patients with rheumatoid arthritis (RA), and to investigate the underlying molecular mechanisms. PBMCs from patients with RA (n=12) and healthy controls (n=10) were cultured to assess osteoclastogenic potential. The number of tartrate­resistant acid phosphatase­positive osteoclasts differentiated from PBMCs isolated from patients with RA was significantly increased compared with that of the healthy controls. In addition, the osteoclast number in patients with RA was correlated with the clinical indicators, Sharp score (r=0.810; P=0.001) and lumbar T­score (r=­0.685; P=0.014). Furthermore, the resorption area was increased in the RA group compared with the healthy controls. The mRNA and protein expression levels in PBMC­derived osteoclasts treated with curcumin were measured by reverse transcription­quantitative polymerase chain reaction and western blotting, respectively. Curcumin inhibited the osteoclastogenic potential of PBMCs, potentially by suppressing activation of extracellular signal­regulated kinases 1 and 2, p38 and c­Jun N­terminal kinase, and inhibiting receptor activator of nuclear factor κB (RANK), c­Fos and nuclear factor of activated T cells (NFATc1) expression. The results of the present study demonstrated that curcumin may inhibit the osteoclastogenic potential of PBMCs from patients with RA through the suppression of the mitogen­activated protein kinase/RANK/c­Fos/NFATc1 signaling pathways, and that curcumin may be a potential novel therapeutic agent for the treatment of bone deterioration in inflammatory diseases such as RA.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Curcumina/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Osteoclastos/patología , Transducción de Señal/efectos de los fármacos , Artritis Reumatoide/inmunología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Leucocitos Mononucleares/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factores de Transcripción NFATC/inmunología , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Proteínas Proto-Oncogénicas c-fos/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología
19.
Immunol Lett ; 171: 5-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26773232

RESUMEN

Antibodies play an important role in therapy and investigative biomedical research. The TNF-family member Receptor Activator of NF-κB (RANK) is known for its role in bone homeostasis and is increasingly recognized as a central player in immune regulation and epithelial cell activation. However, the study of RANK biology has been hampered by missing or insufficient characterization of high affinity tools that recognize RANK. Here, we present a careful description and comparison of two antibodies, RANK-02 obtained by phage display (Newa, 2014 [1]) and R12-31 generated by immunization (Kamijo, 2006 [2]). We found that both antibodies recognized mouse RANK with high affinity, while RANK-02 and R12-31 recognized human RANK with high and lower affinities, respectively. Using a cell apoptosis assay based on stimulation of a RANK:Fas fusion protein, and a cellular NF-κB signaling assay, we showed that R12-31 was agonist for both species. R12-31 interfered little or not at all with the binding of RANKL to RANK, in contrast to RANK-02 that efficiently prevented this interaction. Depending on the assay and species, RANK-02 was either a weak agonist or a partial antagonist of RANK. Both antibodies recognized human Langerhans cells, previously shown to express RANK, while dermal dendritic cells were poorly labeled. In vivo R12-31 agonist activity was demonstrated by its ability to induce the formation of intestinal villous microfold cells in mice. This characterization of two monoclonal antibodies should now allow better evaluation of their application as therapeutic reagents and investigative tools.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Epiteliales/fisiología , Epítopos/metabolismo , Intestinos/efectos de los fármacos , Células de Langerhans/inmunología , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Diferenciación Celular/efectos de los fármacos , Técnicas de Visualización de Superficie Celular , Células Epiteliales/efectos de los fármacos , Epítopos/inmunología , Células HEK293 , Humanos , Inmunización Secundaria , Inmunomodulación , Intestinos/citología , Células Jurkat , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal
20.
Gerontology ; 62(2): 128-37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26088283

RESUMEN

Osteoporosis is a major cause of fractures and associated morbidity in the aged population. The pathogenesis of osteoporosis is multifactorial; whereas traditional pathophysiological concepts emphasize endocrine mechanisms, it has been recognized that also components of the immune system have a significant impact on bone. Since 2000, when the term 'osteoimmunology' was coined, novel insights into the role of inflammatory cytokines by influencing the fine-tuned balance between bone resorption and bone formation have helped to explain the occurrence of osteoporosis in conjunction with chronic inflammatory reactions. Moreover, the phenomenon of a low-grade, chronic, systemic inflammatory state associated with aging has been defined as 'inflamm-aging' by Claudio Franceschi and has been linked to age-related diseases such as osteoporosis. Given the tight anatomical and physiological coexistence of B cells and the bone-forming units in the bone marrow, a role of B cells in osteoimmunological interactions has long been suspected. Recent findings of B cells as active regulators of the RANK/RANKL/OPG axis, of altered RANKL/OPG production by B cells in HIV-associated bone loss or of a modulated expression of genes linked to B-cell biology in response to estrogen deficiency support this assumption. Furthermore, oxidative stress and the generation of advanced glycation end products have emerged as links between inflammation and bone destruction.


Asunto(s)
Linfocitos B/inmunología , Osteoporosis/inmunología , Osteoprotegerina/inmunología , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Citidina Desaminasa/inmunología , Productos Finales de Glicación Avanzada/inmunología , Humanos , Inflamación/inmunología , Estrés Oxidativo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...