Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Int Immunopharmacol ; 126: 111300, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38016346

RESUMEN

Encapsulating peritoneal sclerosis (EPS) is a severe complication of peritoneal dialysis (PD). Surgery is a therapeutic strategy for the treatment of complete intestinal obstruction. However, complete intestinal obstruction in long-term PD results in high mortality and morbidity rates after surgery. Immunopathogenesis participates in EPS formation: CD8, Th1, and Th17 cell numbers increased during the formation of EPS. The anti-inflammatory and immunomodulatory effects of melatonin may have beneficial effects on this EPS. In the present study, we determined that melatonin treatment significantly decreases the Th1 and Th17 cell populations in mice with EPS, decreases the production of IL-1ß, TNF-α, IL-6, and IFN-γ, and increases the production of IL-10. The suppression of Th1 and Th17 cell differentiation by melatonin occurs through the inhibition of dendritic cell (DC) activation by affecting the initiation of the NF-κB signaling pathway in DCs. Our study suggests that melatonin has preventive potential against the formation of EPS in patients with PD.


Asunto(s)
Obstrucción Intestinal , Melatonina , Fibrosis Peritoneal , Humanos , Animales , Ratones , Fibrosis Peritoneal/etiología , FN-kappa B/metabolismo , Melatonina/farmacología , Melatonina/uso terapéutico , Diferenciación Celular , Transducción de Señal , Células Dendríticas/metabolismo , Obstrucción Intestinal/complicaciones , Obstrucción Intestinal/patología
3.
Int J Mol Sci ; 24(8)2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37108686

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease with chronic inflammation, bone erosion, and joint deformation. Synovial tissue in RA patients is full of proinflammatory cytokines and infiltrated immune cells, such as T help (Th) 9, Th17, macrophages, and osteoclasts. Recent reports emphasized a new member of the interleukin (IL)-10 family, IL-26, an inducer of IL-17A that is overexpressed in RA patients. Our previous works found that IL-26 inhibits osteoclastogenesis and conducts monocyte differentiation toward M1 macrophages. In this study, we aimed to clarify the effect of IL-26 on macrophages linking to Th9 and Th17 in IL-9 and IL-17 expression and downstream signal transduction. Murine and human macrophage cell lines and primary culture cells were used and stimulated by IL26. Cytokines expressions were evaluated by flow cytometry. Signal transduction and transcription factors expression were detected by Western blot and real time-PCR. Our results show that IL-26 and IL-9 colocalized in macrophage in RA synovium. IL-26 directly induces macrophage inflammatory cytokines IL-9 and IL-17A expression. IL-26 increases the IL-9 and IL-17A upstream mechanisms IRF4 and RelB expression. Moreover, the AKT-FoxO1 pathway is also activated by IL-26 in IL-9 and IL-17A expressing macrophage. Blockage of AKT phosphorylation enhances IL-26 stimulating IL-9-producing macrophage cells. In conclusion, our results support that IL-26 promotes IL-9- and IL-17-expressing macrophage and might initiate IL-9- and IL-17-related adaptive immunity in rheumatoid arthritis. Targeting IL-26 may a potential therapeutic strategy for rheumatoid arthritis or other IL-9 plus IL-17 dominant diseases.


Asunto(s)
Artritis Reumatoide , Interleucina-17 , Animales , Humanos , Ratones , Artritis Reumatoide/metabolismo , Citocinas/metabolismo , Interleucina-17/genética , Interleucina-17/farmacología , Interleucina-17/metabolismo , Interleucina-9/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Th17 , Interleucinas/farmacología
4.
Ultrason Imaging ; 45(2): 74-84, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36951105

RESUMEN

Breast cancer is the most common form of cancer and is still the second leading cause of death for women in the world. Early detection and treatment of breast cancer can reduce mortality rates. Breast ultrasound is always used to detect and diagnose breast cancer. The accurate breast segmentation and diagnosis as benign or malignant is still a challenging task in the ultrasound image. In this paper, we proposed a classification model as short-ResNet with DC-UNet to solve the segmentation and diagnosis challenge to find the tumor and classify benign or malignant with breast ultrasonic images. The proposed model has a dice coefficient of 83% for segmentation and achieves an accuracy of 90% for classification with breast tumors. In the experiment, we have compared with segmentation task and classification result in different datasets to prove that the proposed model is more general and demonstrates better results. The deep learning model using short-ResNet to classify tumor whether benign or malignant, that combine DC-UNet of segmentation task to assist in improving the classification results.


Asunto(s)
Neoplasias de la Mama , Redes Neurales de la Computación , Femenino , Humanos , Mama/diagnóstico por imagen , Neoplasias de la Mama/diagnóstico por imagen , Ultrasonografía , Ultrasonografía Mamaria , Procesamiento de Imagen Asistido por Computador/métodos
5.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163121

RESUMEN

Type 1 diabetes (T1D) is caused by the destruction of ß cells in pancreatic islets by autoimmune T cells. Islet transplantation has been established as an effective treatment for T1D. However, the survival of islet grafts is often disrupted by recurrent autoimmunity. Alpha-lipoic acid (ALA) has been reported to have immunomodulatory effects and, therefore, may have therapeutic potential in the treatment of T1D. In this study, we investigated the therapeutic potential of ALA in autoimmunity inhibition. We treated non-obese diabetic (NOD) mice with spontaneous diabetes and islet-transplantation mice with ALA. The onset of diabetes was decreased and survival of the islet grafts was extended. The populations of Th1 cells decreased, and regulatory T cells (Tregs) increased in ALA-treated mice. The in vitro Treg differentiation was significantly increased by treatment with ALA. The adoptive transfer of ALA-differentiated Tregs into NOD recipients improved the outcome of the islet grafts. Our results showed that in vivo ALA treatment suppressed spontaneous diabetes and autoimmune recurrence in NOD mice by inhibiting the Th1 immune response and inducing the differentiation of Tregs. Our study also demonstrated the therapeutic potential of ALA in Treg-based cell therapies and islet transplantation used in the treatment of T1D.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/citología , Linfocitos T Reguladores/inmunología , Ácido Tióctico/farmacología , Animales , Antioxidantes/farmacología , Diferenciación Celular , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Supervivencia de Injerto , Ratones , Ratones Endogámicos NOD , Células TH1
6.
Pharmaceuticals (Basel) ; 14(5)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067829

RESUMEN

Type 1 diabetes mellitus (T1D) results from the destruction of insulin-producing ß cells in the islet of the pancreas by lymphocytes. Non-obese diabetic (NOD) mouse is an animal model frequently used for this disease. It has been considered that T1D is a T cell-mediated autoimmune disease. Both CD4+ and CD8+ T cells are highly responsible for the destruction of ß cells within the pancreatic islets of Langerhans. Previous studies have revealed that regulatory T (Treg) cells play a critical role in the homeostasis of the immune system as well as immune tolerance to autoantigens, thereby preventing autoimmunity. Valproic acid (VPA), a branched short-chain fatty acid, is widely used as an antiepileptic drug and a mood stabilizer. Previous reports have demonstrated that VPA treatment decreases the incidence and severity of collagen-induced arthritis and experimental autoimmune neuritis by increasing the population of Treg cells in these mouse disease models. Given the effect of VPA in the induction of Treg cells' population, we evaluated the therapeutic potential and the protective mechanism of VPA treatment in the suppression of graft autoimmune rejection and immune recurrence in syngeneic or allogenic islet transplantation mouse models. In our study, we found that the treatment of VPA increased the expression of forkhead box P3 (FOXP3), which is a critical transcription factor that controls Treg cells' development and function. Our data revealed that 400 mg/kg VPA treatment in recipients effectively prolonged the survival of syngeneic and allogenic islet grafts. The percentage of Treg cells in splenocytes increased in VPA-treated recipients. We also proved that adoptive transfer of VPA-induced Tregs to the transplanted recipients effectively prolonged the survival of islet grafts. The results of this study provide evidence of the therapeutic potential and the underlying mechanism of VPA treatment in syngeneic islet transplantation for T1D. It also provides experimental evidence for cell therapy by adoptive transferring of in vitro VPA-induced Tregs for the suppression of autoimmune recurrence.

7.
Mediators Inflamm ; 2020: 1237281, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32587467

RESUMEN

PURPOSE: Interleukin-1α (IL-1α) is a potent cytokine that plays a role in inflammatory arthritis and bone loss. Decoy receptor 3 (DCR3) is an immune modulator of monocytes and macrophages. The aim of this study was to investigate the mechanism of DCR3 in IL-1α-induced osteoclastogenesis. METHODS: We treated murine macrophages with DCR3 during receptor activator of nuclear factor kappa Β ligand- (RANKL-) plus IL-1α-induced osteoclastogenesis to monitor osteoclast formation by tartrate-resistant acid phosphatase (TRAP) staining. Osteoclast activity was assessed using a pit formation assay. The mechanisms of inhibition were studied by biochemical analyses, including RT-PCR, immunofluorescent staining, flow cytometry, an apoptosis assay, immunoblotting, and ELISA. RESULTS: DCR3 suppresses IL-1α-induced osteoclastogenesis in both primary murine bone marrow-derived macrophages (BMM) and RAW264.7 cells as it inhibits bone resorption. DCR3 induces RANKL-treated osteoclast precursor cells to express IL-1α, secretory IL-1ra (sIL-1ra), intracellular IL-1ra (icIL-1ra), reactive oxygen species (ROS), and Fas ligand and to activate IL-1α-induced interleukin-1 receptor-associated kinase 4 (IRAK4). The suppression of DCR3 during RANKL- or IL-1α-induced osteoclastogenesis may be due to the abundant secretion of IL-1ra, accumulation of ROS, and expression of Fas ligand in apoptotic osteoclast precursor cells. CONCLUSIONS: We concluded that there is an inhibitory effect of DCR3 on osteoclastogenesis via ROS accumulation and ROS-induced Fas ligand, IL-1α, and IL-1ra expression. Our results suggested that the upregulation of DCR3 in preosteoclasts might be a therapeutic target in inflammatory IL-1α-induced bone resorption.


Asunto(s)
Proteína Ligando Fas/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1alfa/metabolismo , Osteoclastos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Miembro 6b de Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Muerte Celular/genética , Muerte Celular/fisiología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Immunoblotting , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Ratones , Osteoclastos/citología , Células RAW 264.7 , Fosfatasa Ácida Tartratorresistente/metabolismo
8.
Cells ; 9(4)2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290250

RESUMEN

Interleukin 26 (IL-26) is a new member of the IL-10 family that is highly expressed in rheumatoid arthritis (RA). However, the functions of IL-26 produced by macrophages in RA have not been elucidated. In the present work, we evaluated the effects and the mechanisms of IL-26 on M1 and M2 macrophage differentiation. Human or mouse macrophage cells were treated with lipopolysaccharides (LPS), interferon gamma (IFNγ), or IL-4 alone or concurrently treated with IL-26 to monitor M1 or M2 macrophage subtypes. The expression level of M1 or M2 macrophage genes was evaluated by reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). The molecular mechanisms of downstream signaling activation during differentiation were investigated by immunoblotting assay. Our results found that IL-26 promoted macrophage cells from CD80+ M1 macrophage differentiation, not from the CD206+ M2 phenotype. The messenger RNA of M1-type macrophage markers tumor necrosis factor alpha (TNFα) and inducible nitric oxide synthase (iNOS) was up-regulated in the IL-26-treated group. Also, the M1-related proinflammatory cytokines TNFα and IL-6 were induced after IL-26 stimulation. Interestingly, IL-10, a cytokine marker of M2 macrophage, was also elevated after IL-26 stimulation. Moreover, the M1-like macrophage stimulated by IL-26 underwent cJUN, nuclear factor kappa B (NF-κB), and signal transducer and activator of transcription 1 (STAT1) activation. Our findings suggested the role of IL-26 in synovial macrophages of active rheumatoid arthritis and provided a new insight into IL-26 as a candidate therapeutic target in rheumatoid arthritis.


Asunto(s)
Interleucinas/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Animales , Diferenciación Celular/fisiología , Polaridad Celular/fisiología , Humanos , Activación de Macrófagos , Macrófagos/citología , Ratones , Fenotipo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Células RAW 264.7
9.
Immunobiology ; 225(3): 151906, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31987604

RESUMEN

Dimethyl sulfoxide (DMSO) was discovered during the 19th century by the German chemical industry. DMSO comprises a highly polar group and two non-polar domains, which render it soluble in both aqueous solutions and organic solutions. Furthermore, DMSO can penetrate the cell membrane of both the mammalian cells and the non-mammalian cells and prevent freeze-thaw injuries to the cells. Thus, it is frequently used for the cryopreservation of cells and tissues for laboratory and clinical applications. In contrast to this traditional application, DMSO has recently been shown to possess immunomodulatory effects, such as immune enhancement, and anti-inflammatory effects in the innate immunity. In addition, DMSO also affects the adaptive immunity by regulating the expression of transcription factors in immune cells. This review briefly summarizes and highlights the roles and immunomodulatory effects of DMSO on the immune system and reveals the future clinical therapeutic potential of DMSO treatment in cancer, in autoimmune diseases and in chronic inflammatory diseases.


Asunto(s)
Dimetilsulfóxido/farmacología , Inmunomodulación/efectos de los fármacos , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Permeabilidad de la Membrana Celular , Crioprotectores , Dimetilsulfóxido/química , Dimetilsulfóxido/uso terapéutico , Humanos , Sistema Inmunológico/diagnóstico por imagen , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata/efectos de los fármacos , Solubilidad
10.
Toxicol Appl Pharmacol ; 378: 114641, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31254568

RESUMEN

Encapsulating peritoneal sclerosis (EPS) is a severe complication of peritoneal dialysis (PD). This disease leads to intestinal obstruction with or without peritonitis. The imbalance between the populations of Th17 and regulatory T (Treg) cells (higher Th17 cells and lower Treg cells) is part of the pathogenesis of EPS formation. We demonstrated that dimethyl sulfoxide (DMSO) effectively inhibited autoimmune diabetes recurrence in the islet transplantation of NOD mice via the induction of the differentiation of Treg cells. In this study, we investigated the therapeutic potential of DMSO in the inhibition of EPS formation by a mouse model. Under DMSO treatment, the thickening of the parietal and visceral peritoneum was significantly reduced. The populations of CD4, CD8, and IFN-γ-producing CD4 and CD8 T cells were decreased. The populations of IL-4-producing CD4 T lymphocytes, IL-10-producing CD4 T lymphocytes, CD4 CD69 T lymphocytes and Treg lymphocytes were increased. The expression levels of the cytokines IFN-γ, IL-17a, TNF-α and IL-23, in ascites, were significantly decreased following the DMSO treatment. Furthermore, the differentiation of Treg cells was induced by DMSO from naïve CD4 T cells in vitro, and these cells were adoptively transferred into the EPS mice and significantly prevented EPS formation, exhibiting a comparable effect to the in vivo DMSO treatment. We also demonstrated that the differentiation of Treg cells by DMSO occurred via the activation of STAT5 by its epigenetic effect, without altering the PI3K-AKT-mTOR or Raf-ERK pathways. Our results demonstrated, for the first time, that in vivo DMSO treatment suppresses EPS formation in a mouse model. Furthermore, the adoptive transfer of Treg cells that were differentiated from naïve CD4 T cells by an in vitro DMSO treatment exhibited a similar effect to the in vivo DMSO treatment for the prevention of EPS formation.


Asunto(s)
Dimetilsulfóxido/inmunología , Fibrosis Peritoneal/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo/métodos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Citocinas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Femenino , Interleucina-17/inmunología , Trasplante de Islotes Pancreáticos/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fosfatidilinositol 3-Quinasas/inmunología , Células Th17/inmunología
11.
Clin Oral Investig ; 21(1): 419-427, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27073100

RESUMEN

OBJECTIVES: Advanced oral cancer is a major public health concern because of a lack of effective prevention and treatment. Triptolide (TPL), a diterpenoid triepoxide derived from the Chinese herb Tripterygium wilfordii, has been demonstrated to possess strong anticancer properties. In this study, we investigated whether TPL exerts anticancer effects on the tumor microenvironment of head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS: Human macrophage-like U937 cells were co-inoculated with oral cancer SAS cells in a noncontact transwell coculture system. Cytokine expression was detected using ELISA, and cell proliferation was detected using methylene blue. RNA levels were detected using qPCR. Protein levels were detected using Western blot analysis. In vivo experiments involved using xenografted NOD/SCID mice. RESULTS: Our results demonstrated that TPL inhibited the growth of SAS cells co-inoculated with U937 cells in vitro and in vivo. TPL inhibited the invasion, migration ability, and angiogenesis of SAS cells co-inoculated with U937 cells. Expression of cytokines IL-6, IL-8, and TNF-α was induced by co-inoculation, but TPL repressed their expression. CONCLUSION: TPL suppressed the expression of cytokines IL-6, IL-8, and TNF-α, as well as tumor growth, invasion, migration, and angiogenesis in the co-inoculation of human tongue cancer cells with macrophage-like U937 cells. CLINICAL RELEVANCE: TPL is a potential candidate among novel chemotherapeutic agents or adjuvants for modulating tumor-associated macrophages in a tumor microenvironment of HNSCC.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diterpenos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Fenantrenos/farmacología , Células U937 , Animales , Western Blotting , Carcinoma de Células Escamosas/tratamiento farmacológico , Técnicas de Cocultivo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Compuestos Epoxi/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Reacción en Cadena de la Polimerasa , Microambiente Tumoral
12.
Rheumatology (Oxford) ; 55(11): 2074-2083, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27550297

RESUMEN

OBJECTIVE: IL-26 has been shown to have high expression in RA. However, the effects of IL-26 on bone destruction in RA have not been evaluated. The aim of this study was to investigate the effects and mechanisms of IL-26 on RANK ligand (RANKL)-induced osteoclastogenesis. METHODS: We treated cells with IL-26 in RANKL-induced oseteoclastogenesis to monitor osteoclast formation by tartrate-resistant acid phosphatase (TRAP) staining. Osteoclast activity was assessed by pit formation assay and F-actin ring formation. The mechanism of the inhibition was studied by biochemical analyses such as RT-PCR, immunofluorescence staining and immunoblotting. In addition, cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. RESULTS: IL-26 inhibited RANKL-induced TRAP-positive multinucleated cells and inhibited RANKL-induced nuclear factor κB (NF-κB) activation and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) nuclear translocation in RAW264.7 cells. Also, IL-26 significantly inhibited the bone-resorbing activity and F-actin ring formation ability of mature osteoclasts. Moreover, IL-26 suppressed RANKL-induced mitogen-activated protein kinase activation and NFATc1 downstream gene expression. CONCLUSION: We suggest that the inhibitory activity of IL-26 on osteoclastogenesis is via down-regulation of RANKL-induced NF-κB and NFATc1 expression. Our results suggest IL-26 as a possible new remedy against osteolytic bone destruction.


Asunto(s)
Interleucinas/fisiología , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteogénesis/fisiología , Animales , Artritis Reumatoide/fisiopatología , Resorción Ósea/fisiopatología , Regulación hacia Abajo , Humanos , Ratones , Osteoclastos/fisiología , Osteólisis/fisiopatología , Ligando RANK/metabolismo , Células RAW 264.7
13.
Int Immunopharmacol ; 31: 169-77, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26735612

RESUMEN

Melatonin is the major product secreted by the pineal gland at night and displays multifunctional properties, including immunomodulatory functions. In this study, we investigated the therapeutic effect of melatonin in experimental autoimmune encephalomyelitis (EAE). We demonstrated that melatonin exhibits a therapeutic role by ameliorating the clinical severity and restricting the infiltration of inflammatory Th17 cells into the CNS of mice with myelin oligodendrocyte glycoprotein (MOG)-induced EAE. Furthermore, melatonin enhances splenic interleukin (IL)-10 expression in regulatory T cells by inducing IL-27 expression in the splenic DC; it also suppresses the expression of IFN-γ, IL-17, IL-6, and CCL20 in the CNS and inhibits antigen-specific T cell proliferation. However, there were no significant differences in the percentage of splenic regulatory T cells. These data provide the first evidence that the therapeutic administration of melatonin is effective in mice with EAE and modulates adaptive immunity centrally and peripherally. Thus, we suggest that melatonin could play an adjunct therapeutic role in treating human CNS autoimmune diseases such as multiple sclerosis. Melatonin merits further studies in animals and humans.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Interleucina-10/metabolismo , Melatonina/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Regulación hacia Arriba/efectos de los fármacos
14.
Clin Oral Investig ; 20(3): 533-40, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26205068

RESUMEN

OBJECTIVES: Death domain-associated protein (Daxx) has been recently implicated as a positive factor in ovarian cancer and prostate cancer, but the role of Daxx in oral squamous cell carcinoma (OSCC) has never been addressed. Herein, we investigate the expression and function of Daxx in OSCC. MATERIALS AND METHODS: RT-quantitative PCR, Western blotting, and immunohistochemistry were used to evaluation of the expression of Daxx in human OSCC cell lines and clinical surgical specimens. Short hairpin RNA targeting Daxx was transduced by lentivirus infection to knockdown the expression of Daxx in SAS and SCC25 cell lines, and the influence of this knockdown was evaluated by analyzing the growth and the cell cycle in transduced cells. Immunoprecipitation and sequential chromatin immunoprecipitation-quantitative PCR were used to analyze the associations between Daxx, TCF4, and cyclin D1 promoter. Xenograft tumor model was used to evaluate the in vivo tumorigenicity of Daxx in OSCC. RESULTS: Daxx mRNA and protein expression are elevated in several OSCC cell lines and human OSCC samples in comparison to those in normal tissue. We further find that depletion of Daxx decreases OSCC cell growth activity through G1 cell cycle arrest. Daxx silencing reduces cyclin D1 expression via a Daxx-TCF4 interaction, whereas the Daxx depletion-mediated G1 arrest can be relieved by ectopic expression of cyclin D1. Moreover, we show that in OSCC clinical samples, the expression of Daxx is significantly correlated with that of cyclin D1. CONCLUSION: Our data demonstrate the importance of Daxx in regulation of cyclin D1 expression and provide the first evidence that Daxx exhibits tumor-promoting activity in OSCC. CLINICAL RELEVANCE: Daxx plays an important role in malignant transformation of OSCC and may serves as a target for cancer prevention and treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Carcinoma de Células Escamosas/metabolismo , Ciclina D1/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas Nucleares/fisiología , Factores de Transcripción/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Carcinoma de Células Escamosas/patología , Ciclo Celular , Línea Celular Tumoral , Proteínas Co-Represoras , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Masculino , Ratones , Persona de Mediana Edad , Chaperonas Moleculares , Neoplasias de la Boca/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción 4
15.
J Biol Chem ; 290(49): 29329-44, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26468284

RESUMEN

Glucosamine has immunomodulatory effects on autoimmune diseases. However, the mechanism(s) through which glucosamine modulates different T cell subsets and diseases remain unclear. We demonstrate that glucosamine impedes Th1, Th2, and iTreg but promotes Th17 differentiation through down-regulating N-linked glycosylation of CD25 and subsequently inhibiting its downstream Stat5 signaling in a dose-dependent manner. The effect of glucosamine on T helper cell differentiation was similar to that induced by anti-IL-2 treatment, further supporting an IL-2 signaling-dependent modulation. Interestingly, excess glucose rescued this glucosamine-mediated regulation, suggesting a functional competition between glucose and glucosamine. High-dose glucosamine significantly decreased Glut1 N-glycosylation in Th1-polarized cells. This finding suggests that both down-regulated IL-2 signaling and Glut1-dependent glycolytic metabolism contribute to the inhibition of Th1 differentiation by glucosamine. Finally, glucosamine treatment inhibited Th1 cells in vivo, prolonged the survival of islet grafts in diabetic recipients, and exacerbated the severity of EAE. Taken together, our results indicate that glucosamine interferes with N-glycosylation of CD25, and thereby attenuates IL-2 downstream signaling. These effects suggest that glucosamine may be an important modulator of T cell differentiation and immune homeostasis.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Glucosamina/química , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Animales , Enfermedades Autoinmunes/metabolismo , Regulación hacia Abajo , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Glicosilación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Transducción de Señal , Células TH1/citología , Células Th17/citología , Células Th2/citología
16.
Toxicol Appl Pharmacol ; 282(2): 207-14, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25481496

RESUMEN

Type 1 diabetes mellitus (T1D) is caused by the destruction of insulin-producing ß cells in pancreatic islets by autoimmune T cells. Islet transplantation has been established as an effective therapeutic strategy for T1D. However, the survival of islet grafts can be disrupted by recurrent autoimmunity. Dimethyl sulfoxide (DMSO) is a solvent for organic and inorganic substances and an organ-conserving agent used in solid organ transplantations. DMSO also exerts anti-inflammatory, reactive oxygen species scavenger and immunomodulatory effects and therefore exhibits therapeutic potential for the treatment of several human inflammatory diseases. In this study, we investigated the therapeutic potential of DMSO in the inhibition of autoimmunity. We treated an animal model of islet transplantation (NOD mice) with DMSO. The survival of the syngeneic islet grafts was significantly prolonged. The population numbers of CD8, DC and Th1 cells were decreased, and regulatory T (Treg) cell numbers were increased in recipients. The expression levels of IFN-γ and proliferation of T cells were also reduced following DMSO treatment. Furthermore, the differentiation of Treg cells from naive CD4 T cells was significantly increased in the in vitro study. Our results demonstrate for the first time that in vivo DMSO treatment suppresses spontaneous diabetes and autoimmune recurrence in NOD mice by inhibiting the Th1 immune response and inducing the differentiation of Treg cells.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Diabetes Mellitus/prevención & control , Dimetilsulfóxido/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Insulina/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Ratones SCID
17.
PLoS One ; 9(8): e104703, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25127132

RESUMEN

OBJECTIVE: Oral squamous cell carcinoma (OSCC) is a prevalent cancer, especially in developing countries. Anthracyclines and their anthraquinone derivatives, such as doxorubicin, exhibit a cell growth inhibitory effect and have been used as anti-cancer drugs for many years. However, the cardiotoxicity of anthracycline antibiotics is a major concern in their clinical application. NSC745885 is a novel compound synthesized from 1,2-diaminoanthraquinone, which subsequently reacts with thionyl chloride and triethylamine. The present study aimed to investigate the anti-oral cancer potential and the safety of NSC745885. METHODS: We investigated the anti-cancer potential of NSC745885 in oral squamous carcinoma cell lines and in an in vivo oral cancer xenograft mouse model. The expression of apoptotic related genes were evaluated by real-time RT-PCR and western bloting, and the in vivo assessment of apoptotic marker were measured by immunohistochemical staining. The anti-tumor efficiency and safety between doxorubicin and NSC745885 were also compared. RESULTS: Our results demonstrated that NSC745885 exhibits anti-oral cancer activity through the induction of apoptosis in cancer cells and in tumor-bearing mice, and this treatment did not induce marked toxicity in experimental mice. This compound also exhibits a comparable anti-tumor efficiency and a higher safety in experimental mice when compared to doxorubicin. CONCLUSIONS: The data of this study provide evidence for NSC745885 as a potential novel therapeutic drug for the treatment of human OSCC.


Asunto(s)
Antraquinonas/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Tiadiazoles/uso terapéutico , Neoplasias de la Lengua/tratamiento farmacológico , Animales , Antraquinonas/efectos adversos , Antraquinonas/química , Antineoplásicos/uso terapéutico , Caspasa 3/biosíntesis , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Recurrencia Local de Neoplasia/tratamiento farmacológico , ARN Mensajero/biosíntesis , Tiadiazoles/efectos adversos , Lengua/patología , Proteína Inhibidora de la Apoptosis Ligada a X/biosíntesis , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J BUON ; 19(2): 419-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24965401

RESUMEN

PURPOSE: Primary neuroendocrine carcinoma of the breast (NECB) is a rare distinct type of breast carcinoma. There are only some case reports on this topic published in the past. There is still little known on the optimal treatment outcomes, while a wide variety of treatments is proposed by several authors. In this study we searched the literature on NECB in PubMed to clarify its prognosis and possible optimal therapeutic strategies. METHODS: Eighty-six cases of primary NEC, included our case, were collected from PubMed between 1980 and 2013. Initial stage, estrogen receptor (ER)/progesterone receptor (PR)/ human epidermal growth factor receptor 2 (HER-2), surgical procedures, adjuvant treatment and overall survive (OS) were analyzed using the Statistical Package for the Social Sciences ( SPSS, v 16.0 ). RESULTS: All 86 patients enrolled were eligible. Their mean age at diagnosis was 53.9 years (range 25-83) and 1 case was in a male. Overall survival (OS) at 48 months was 83.5%. Patients with enlarged tumor size (10 patients with tumor size >5.0 cm) or advanced stage (stage III 15 patients, stage IV 2 patients) had poor OS (48-month OS: 51.4 vs 97.1% with tumors >5cm vs ≤2cm, respectively and 0.0%, 68.1%, 72.9% and 95.8% in stage IV, III, II and I, respectively). Patients with positive ER, PR or HER-2 had significantly better OS than did those without (ER, p<0.001; PR, p<0.001; HER-2, p=0.082). Besides, all 60 patients with initial primary surgery and without lymph node dissection (LND) showed better OS than those with initial primary surgery without LND, the difference however being not significant (p=0.133). CONCLUSION: Definite diagnosis and clinical stage are prerequisites in the initial approach in NECB. When detected early the disease may have a good prognosis with combined modality treatment such as chemotherapy, surgery, and radiation therapy. An appropriate therapeutic strategy for this group is also important. Our analysis showed that for patients with early localized disease only primary surgery is recommended and LND is optional. In patients with positive steroid receptors postoperative hormonotherapy is suggested.


Asunto(s)
Neoplasias de la Mama Masculina/patología , Neoplasias de la Mama/patología , Carcinoma Neuroendocrino/patología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Neoplasias de la Mama Masculina/mortalidad , Neoplasias de la Mama Masculina/terapia , Carcinoma Neuroendocrino/mortalidad , Carcinoma Neuroendocrino/terapia , Femenino , Humanos , Escisión del Ganglio Linfático , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias
19.
Planta Med ; 80(4): 255-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24510366

RESUMEN

Advanced oral cancer has a poor prognosis because of the lack of an effective treatment. We explored the efficiency of combined treatment with triptolide and ionizing radiation for treating oral cancer. Human tongue cancer cells were treated with triptolide, ionizing radiation, or triptolide plus ionizing radiation. Cell proliferation, cell cycle arrest, and apoptotic influences were analyzed by FACS and immunohistochemistry. Tumor potency was examined in an in vivo human tongue cancer cells xenograft mouse model. Our results demonstrated that triptolide caused a marked reduction in colony number that was further enhanced with increasing doses of ionizing radiation. Triptolide increased apoptosis and decreased the expression of anti-apoptotic proteins. In vivo, combination treatment synergistically reduced tumor weight and volume possibly via the induction of apoptosis and reduction in anti-apoptotic protein expression. In conclusion, triptolide plus ionizing radiation treatment had synergistic anti-tumor effects, especially in vivo, and may be a promising combined modality therapy for advanced oral cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Diterpenos/uso terapéutico , Fenantrenos/uso terapéutico , Fitoterapia , Neoplasias de la Lengua/tratamiento farmacológico , Tripterygium/química , Animales , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Quimioterapia Adyuvante , Diterpenos/farmacología , Compuestos Epoxi/farmacología , Compuestos Epoxi/uso terapéutico , Humanos , Ratones , Fenantrenos/farmacología , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Lengua/efectos de los fármacos , Neoplasias de la Lengua/radioterapia , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Mol Sci ; 14(6): 11742-66, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23727938

RESUMEN

Melatonin is the major secretory product of the pineal gland during the night and has multiple activities including the regulation of circadian and seasonal rhythms, and antioxidant and anti-inflammatory effects. It also possesses the ability to modulate immune responses by regulation of the T helper 1/2 balance and cytokine production. Autoimmune diseases, which result from the activation of immune cells by autoantigens released from normal tissues, affect around 5% of the population. Activation of autoantigen-specific immune cells leads to subsequent damage of target tissues by these activated cells. Melatonin therapy has been investigated in several animal models of autoimmune disease, where it has a beneficial effect in a number of models excepting rheumatoid arthritis, and has been evaluated in clinical autoimmune diseases including rheumatoid arthritis and ulcerative colitis. This review summarizes and highlights the role and the modulatory effects of melatonin in several inflammatory autoimmune diseases including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes mellitus, and inflammatory bowel disease.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/etiología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Melatonina/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Melatonina/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...