Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Med Oncol ; 39(8): 117, 2022 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-35674832

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common malignancy in children and relapsed B-ALL is the leading cause of mortality in children with leukemia due to a lack of response to treatment. S100A8 is a low molecular weight calcium-binding intracellular protein that is expressed in certain cells, and its increased expression is seen in most tumors as well as in relapsed childhood B-ALL cases. The present study indicates the important role of S100A8 in improving viability and resistance to chemotherapy in relapsed B-ALL lymphoblasts. S100A8 levels were compared in B-ALL and relapsed B-ALL lymphoblasts that were sensitive and resistant to Vincristine, respectively. S100A8 was inhibited in the lymphoblasts of two patients by antisense locked nucleic acid (LNA) GapmeRs and the decreased expression of S100A8 was evaluated using quantitative real-time PCR and ELISA. Then, the S100A8 antisense LNA GapmeRs-transfected cells were treated with Vincristine and the expression levels of S100A8 mRNA and S100A8 protein were re-determined. At all of these stages, cell viability and LC50 were assessed by MTT assay. The results showed that S100A8 levels in relapsed B-ALL lymphoblasts were significantly higher than B-ALL lymphoblasts. Moreover, the increase in S100A8 expression was proportionate to the increase in Vincristine resistance in these cells. The S100A8 knockdown procedure using antisense LNA GapmeRs decreased the cell viability and increased vincristine sensitivity in lymphoblasts of two patients, and it also increased the sensitivity to chemotherapy in relapsed B-ALL lymphoblasts. According to the findings of the present study, S100A8 is effective in developing lymphoblast resistance to chemotherapy, and its enhanced expression may contribute to shifting B-ALL into the relapse phase of the illness. As a result, S100A8 may be a valuable target for managing and improving relapses B-ALL.


Asunto(s)
Calgranulina A , Leucemia-Linfoma Linfoblástico de Células Precursoras , Calgranulina A/antagonistas & inhibidores , Calgranulina A/genética , Niño , Humanos , Linfocitos/patología , Oligonucleótidos , Oligonucleótidos Antisentido , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Recurrencia , Vincristina/farmacología
2.
Molecules ; 26(5)2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33801279

RESUMEN

Deregulations of the expression of the S100A8 and S100A9 genes and/or proteins, as well as changes in their plasma levels or their levels of secretion in the bone marrow microenvironment, are frequently observed in acute myeloblastic leukemias (AML) and acute lymphoblastic leukemias (ALL). These deregulations impact the prognosis of patients through various mechanisms of cellular or extracellular regulation of the viability of leukemic cells. In particular, S100A8 and S100A9 in monomeric, homodimeric, or heterodimeric forms are able to modulate the survival and the sensitivity to chemotherapy of leukemic clones through their action on the regulation of intracellular calcium, on oxidative stress, on the activation of apoptosis, and thanks to their implications, on cell death regulation by autophagy and pyroptosis. Moreover, biologic effects of S100A8/9 via both TLR4 and RAGE on hematopoietic stem cells contribute to the selection and expansion of leukemic clones by excretion of proinflammatory cytokines and/or immune regulation. Hence, the therapeutic targeting of S100A8 and S100A9 appears to be a promising way to improve treatment efficiency in acute leukemias.


Asunto(s)
Antineoplásicos/farmacología , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Leucemia Mieloide Aguda/patología , Terapia Molecular Dirigida , Animales , Calgranulina A/antagonistas & inhibidores , Calgranulina B/química , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/metabolismo , Transducción de Señal
3.
J Mol Med (Berl) ; 99(1): 131-145, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33169236

RESUMEN

In patients with interstitial pneumonia, pulmonary fibrosis is an irreversible condition that can cause respiratory failure. Novel treatments for pulmonary fibrosis are necessary. Inflammation is thought to activate lung fibroblasts, resulting in pulmonary fibrosis. Of the known inflammatory molecules, we have focused on S100A8/A9 from the onset of inflammation to the subsequent progression of inflammation. Our findings confirmed the high expression of S100A8/A9 in specimens from patients with pulmonary fibrosis. An active role of S100A8/A9 was demonstrated not only in the proliferation of fibroblasts but also in the fibroblasts' differentiation to myofibroblasts (the active form of fibroblasts). S100A8/A9 also forced fibroblasts to upregulate the production of collagen. These effects were induced via the receptor of S100A8/A9, i.e., the receptor for advanced glycation end products (RAGE), on fibroblasts. The anti-S100A8/A9 neutralizing antibody inhibited the effects of S100A8/A9 on fibroblasts and suppressed the progression of fibrosis in bleomycin (BLM)-induced pulmonary fibrosis mouse model. Our findings strongly suggest a crucial role of S100A8/A9 in pulmonary fibrosis and the usefulness of S100A8/A9-targeting therapy for fibrosis interstitial pneumonia. HIGHLIGHTS: S100A8/A9 level is highly upregulated in the IPF patients' lungs as well as the blood. S100A8/A9 promotes not only the growth of fibroblasts but also differentiation to myofibroblasts. The cell surface RAGE acts as a crucial receptor to the extracellular S100A8/A9 in fibroblasts. The anti-S100A8/A9 antibody effectively suppresses the progression of IPF in a mouse model. In idiopathic pulmonary fibrosis (IPF), S100A8/A9, a heterodimer composed of S100A8 and S100A9 proteins, plays a crucial role in the onset of inflammation and the subsequent formation of a feed-forward inflammatory loop that promotes fibrosis. (1) The local, pronounced increase in S100A8/A9 in the injured inflammatory lung region-which is provided mainly by the activated neutrophils and macrophages-exerts strong inflammatory signals accompanied by dozens of inflammatory soluble factors including cytokines, chemokines, and growth factors that further act to produce and secrete S100A8/A9, eventually making a sustainable inflammatory circuit that supplies an indefinite presence of S100A8/A9 in the extracellular space with a mal-increased level. (2) The elevated S100A8/A9 compels fibroblasts to activate through receptor for advanced glycation end products (RAGE), one of the major S100A8/A9 receptors, resulting in the activation of NFκB, leading to fibroblast mal-events (e.g., elevated cell proliferation and transdifferentiation to myofibroblasts) that actively produce not only inflammatory cytokines but also collagen matrices. (3) Finally, the S100A8/A9-derived activation of lung fibroblasts under a chronic inflammation state leads to fibrosis events and constantly worsens fibrosis in the lung. Taken together, these findings suggest that the extracellular S100A8/A9 heterodimer protein is a novel mainstay soluble factor for IPF that exerts many functions as described above (1-3). Against this background, we herein applied the developed S100A8/A9 neutralizing antibody to prevent IPF. The IPF imitating lung fibrosis in an IPF mouse model was effectively blocked by treatment with the antibody, leading to enhanced survival. The developed S100A8/A9 antibody, as an innovative novel biologic, may help shed light on the difficulties encountered with IPF therapy in clinical settings.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/uso terapéutico , Bleomicina , Calgranulina A/antagonistas & inhibidores , Calgranulina A/sangre , Calgranulina B/sangre , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Fibrosis , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/metabolismo , Regulación hacia Arriba , Adulto Joven
4.
Clin Pharmacol Ther ; 106(6): 1198-1208, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31544235

RESUMEN

Intensive research over the last 3 decades has unequivocally demonstrated the relevance of inflammation in heart failure (HF). Despite our current and ever increasing knowledge about inflammation, the clinical success of antiinflammatory and immunomodulatory therapies in HF is still limited. This review outlines the complexity and diversity of inflammation, its reciprocal interaction with HF, and addresses future perspectives, calling for immunomodulatory therapies that are specific for factors that activate the immune system without the risk of nonspecific immune suppression.


Asunto(s)
Antiinflamatorios/uso terapéutico , Insuficiencia Cardíaca/terapia , Factores Inmunológicos/uso terapéutico , Antagonistas Adrenérgicos beta/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Aterosclerosis/inmunología , Calgranulina A/antagonistas & inhibidores , Calgranulina B , Citocinas/inmunología , Desarrollo de Medicamentos , Insuficiencia Cardíaca/inmunología , Humanos , Inflamación/inmunología , Trasplante de Células Madre Mesenquimatosas , Metotrexato/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Linfocitos T Reguladores , Inhibidores del Factor de Necrosis Tumoral/efectos adversos , Factor de Necrosis Tumoral alfa/inmunología
5.
Eur Heart J ; 40(32): 2713-2723, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31292614

RESUMEN

AIMS: Neutrophils have both detrimental and beneficial effects in myocardial infarction (MI), but little is known about the underlying pathways. S100A8/A9 is a pro-inflammatory alarmin abundantly expressed in neutrophils that is rapidly released in the myocardium and circulation after myocardial ischaemia. We investigated the role of S100A8/A9 in the innate immune response to MI. METHODS AND RESULTS: In 524 patients with acute coronary syndrome (ACS), we found that high plasma S100A8/A9 at the time of the acute event was associated with lower left ventricular ejection fraction (EF) at 1-year and increased hospitalization for heart failure (HF) during follow-up. In wild-type C57BL/6 mice with MI induced by permanent coronary artery ligation, treatment with the S100A9 blocker ABR-238901 during the inflammatory phase of the immune response inhibited haematopoietic stem cell proliferation and myeloid cell egression from the bone marrow. The treatment reduced the numbers of neutrophils and monocytes/macrophages in the myocardium, promoted an anti-inflammatory environment, and significantly improved cardiac function compared with MI controls. To mimic the clinical scenario, we further confirmed the effects of the treatment in a mouse model of ischaemia/reperfusion. Compared with untreated mice, 3-day ABR-238901 treatment significantly improved left ventricular EF (48% vs. 35%, P = 0.002) and cardiac output (15.7 vs. 11.1 mL/min, P = 0.002) by Day 21 post-MI. CONCLUSION: Short-term S100A9 blockade inhibits inflammation and improves cardiac function in murine models of MI. As an excessive S100A8/A9 release is linked to incident HF, S100A9 blockade might represent a feasible strategy to improve prognosis in ACS patients.


Asunto(s)
Calgranulina B/metabolismo , Inflamación/metabolismo , Células Mieloides/metabolismo , Infarto del Miocardio/metabolismo , Animales , Calgranulina A/antagonistas & inhibidores , Calgranulina A/sangre , Calgranulina A/metabolismo , Calgranulina B/sangre , Fármacos Cardiovasculares/farmacología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Células Mieloides/efectos de los fármacos , Miocardio/metabolismo
6.
J Neuroinflammation ; 15(1): 252, 2018 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-30180864

RESUMEN

BACKGROUND: Depression is one of the most common mental disorders characterized mainly by low mood and loss of interest or pleasure. About a third of patients with depression do not respond to classic antidepressant treatments. Recent evidence suggests that Mrp8/14 (myeloid-related protein 8/14) plays a crucial role in cognitive dysfunction and neuroinflammatory diseases, yet its role in mood regulation remains largely uninvestigated. In the present work, we explored the potential role of Mrp8/14 in the progression of depression. METHODS: After 4 weeks of chronic unpredictable mild stress (CUMS), depressive-like symptoms and Mrp8/14 were determined. To verify the effects of Mrp8/14 on depressive-like behaviors, the inhibitor TAK-242 and recombinant Mrp8/14 were used. Furthermore, the molecular mechanisms in Mrp8/14-induced behavioral and biological changes were examined in vivo and ex vivo. RESULTS: Four-week CUMS contributed to the development of depressive symptoms. Mrp8 and Mrp14 were upregulated in the hippocampus and serum after exposure to CUMS. Pharmacological inhibition of Mrp14 attenuated CUMS-induced TLR4/NF-κB signaling activation and depressive-like behaviors. Furthermore, central administration of recombinant Mrp8, Mrp14, and Mrp8/14 resulted in neuroinflammation and depressive-like behaviors. Mrp8/14-provoked proinflammatory effects and depressive-like behaviors were improved by pretreatment with a TLR4 inhibitor. Moreover, pharmacological inhibition of TLR4 reduced the release of nitric oxide and reactive oxygen species in Mrp8/14-activated BV2 microglia. CONCLUSIONS: These data suggest that the hippocampal Mrp8/14-TLR4-mediated neuroinflammation contributes to the development of depressive-like behaviors. Targeting the Mrp8/14 may be a novel promising antidepressant approach.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Depresión/patología , Regulación de la Expresión Génica/fisiología , Hipocampo/metabolismo , Animales , Calgranulina A/antagonistas & inhibidores , Línea Celular Transformada , Citocinas/metabolismo , Depresión/tratamiento farmacológico , Depresión/etiología , Modelos Animales de Enfermedad , Preferencias Alimentarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Inmunosupresores/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/metabolismo , Quinolinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estrés Psicológico/complicaciones , Sacarosa/administración & dosificación , Sulfonamidas/farmacología
7.
Front Immunol ; 9: 1298, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942307

RESUMEN

S100A8 and S100A9 (also known as MRP8 and MRP14, respectively) are Ca2+ binding proteins belonging to the S100 family. They often exist in the form of heterodimer, while homodimer exists very little because of the stability. S100A8/A9 is constitutively expressed in neutrophils and monocytes as a Ca2+ sensor, participating in cytoskeleton rearrangement and arachidonic acid metabolism. During inflammation, S100A8/A9 is released actively and exerts a critical role in modulating the inflammatory response by stimulating leukocyte recruitment and inducing cytokine secretion. S100A8/A9 serves as a candidate biomarker for diagnosis and follow-up as well as a predictive indicator of therapeutic responses to inflammation-associated diseases. As blockade of S100A8/A9 activity using small-molecule inhibitors or antibodies improves pathological conditions in murine models, the heterodimer has potential as a therapeutic target. In this review, we provide a comprehensive and detailed overview of the distribution and biological functions of S100A8/A9 and highlight its application as a diagnostic and therapeutic target in inflammation-associated diseases.


Asunto(s)
Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Animales , Autoinmunidad , Biomarcadores , Calgranulina A/antagonistas & inhibidores , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunidad , Inflamación/tratamiento farmacológico , Inflamación/patología , Terapia Molecular Dirigida , Transporte de Proteínas
8.
Med Sci Monit ; 24: 1836-1846, 2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-29595187

RESUMEN

BACKGROUND The aim of this study was to investigate the expression and silencing of the S100A8 gene, which encodes the S100 calcium-binding protein A8 (S100A8), and apoptosis and phosphorylation of protein kinase B (Akt) in tissue samples of endometrial carcinoma and HEC-1A endometrial adenocarcinoma cells in vitro. MATERIAL AND METHODS Immunohistochemistry (IHC) was used to detect expression of the S100A8 protein in 74 tissue samples of endometrial cancer and 22 normal endometrial tissue samples. A stable S100A8 gene knockdown cell line was constructed using lentiviral packing short hairpin RNA (shRNA) transfected into HEC-1A cells. S100A8 mRNA and S100A8 protein levels were detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting. The effects of expression of the S100A8 gene by endometrial cancer cells was investigated by the MTT assay, cell cycle and apoptotic assays, qRT-PCR, and Western blotting. RESULTS IHC showed high levels of expression of S100A8 protein in endometrial carcinoma tissues, and HEC-1A adenocarcinoma cells (in G1 and G2). Increased expression of S100A8 protein was found endometrial cancer tissues compared with normal endometrial tissues (79.7% vs. 4.5%). S100A8 gene knockdown reduced cell proliferation in the HEC-1A cells compared with control cells, induced cell apoptosis, inhibited the phosphorylation of protein kinase B (Akt), and induced the expression of pro-apoptotic genes, including the cytochrome C gene, CYCS, BAD, BAX, FOXO1, FOXO3, CASP9, and CASP3. CONCLUSIONS In endometrial carcinoma cells, down-regulation of the S100A8 gene induced cell apoptosis via inhibition of the phosphorylated or active form of protein kinase B (Akt).


Asunto(s)
Calgranulina A/genética , Neoplasias Endometriales/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Apoptosis/efectos de los fármacos , Calgranulina A/antagonistas & inhibidores , Calgranulina A/biosíntesis , Ciclo Celular/fisiología , División Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Regulación hacia Abajo , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Fosforilación , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética
9.
Pharmacol Ther ; 167: 120-131, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27492899

RESUMEN

Neutrophils and monocytes belong to the first line of immune defence cells and are recruited to sites of inflammation during infection or sterile injury. Both cells contain huge amounts of the heterodimeric protein S100A8/A9 in their cytoplasm. S100A8/A9 belongs to the Ca2+ binding S100 protein family and has recently gained a lot of interest as a critical alarmin modulating the inflammatory response after its release (extracellular S100A8/A9) from neutrophils and monocytes. Extracellular S100A8/A9 interacts with the pattern recognition receptors Toll-like receptor 4 (TLR4) and Receptor for Advanced Glycation Endproducts (RAGE) promoting cell activation and recruitment. Besides its biological function, S100A8/A9 (also known as myeloid related protein 8/14, MRP8/14) was identified as interesting biomarker to monitor disease activity in chronic inflammatory disorders including inflammatory bowel disease and rheumatoid arthritis. Furthermore, S100A8/A9 has been tested successfully in pre-clinical imaging studies to localize sites of infection or sterile injury. Finally, recent evidence using small molecule inhibitors for S100A8/A9 also suggests that blocking S100A8/A9 activity exerts beneficial effects on disease activity in animal models of autoimmune diseases including multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis and inflammatory bowel disease. This review will provide a comprehensive and detailed overview into the structure and biological function of S100A8/A9 and also will give an outlook in terms of diagnostic and therapeutic applications targeting S100A8/A9.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Inflamación/tratamiento farmacológico , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Calcio/metabolismo , Calgranulina A/antagonistas & inhibidores , Calgranulina B/efectos de los fármacos , Humanos , Inflamación/inmunología , Inflamación/patología , Terapia Molecular Dirigida , Monocitos/metabolismo , Neutrófilos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor Toll-Like 4/metabolismo
10.
Oncogene ; 35(11): 1445-56, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26165843

RESUMEN

S100A8/A9 is a major component of the acute phase of inflammation, and appears to regulate cell proliferation, redox regulation and chemotaxis. We previously reported that S100A8/S100A9 are upregulated in the premetastatic lung. However, the detailed mechanisms by which S100A8 contributes to tumor progression have not been elucidated. In this study, we investigated the TLR4/MD-2 dependency by S100A8 on tumor progression. We found that S100A8 (2-89) peptide stimulated cell migration in a manner dependent on TLR4, MD-2 and MyD88. The S100A8 (2-89) peptide also activated p38 and NF-κB in TLR4-dependent manner. The peptide induced the upregulation of both IL-6 and Ccl2 in peritoneal macrophages obtained from wild-type mice, but not TLR4-deficient mice. We then investigated the responsible region of S100A8 for TLR4/MD-2 binding by a binding assay, and found that C-terminal region of S100A8 binds to TLR4/MD-2 complex. To further evaluate the TLR4 dependency on tumor microenvironment, Lewis lung carcinoma-bearing mice were treated with Eritoran, an antagonist of TLR4/MD-2 complex. We found that both tumor volume and pulmonary recruitment of myeloid-derived suppressor cells were reduced with the treatment of Eritoran for five consecutive days. Eritoran reduced the development of tumor vasculature, and increased tumor-infiltration of CD8(+) T-cells. Taken together, S100A8 appears to play a crucial role in the activation of the TLR4/MD-2 pathway and the promotion of a tumor growth-enhancing immune microenvironment.


Asunto(s)
Calgranulina A/antagonistas & inhibidores , Carcinoma Pulmonar de Lewis/inmunología , Disacáridos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Fosfatos de Azúcar/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Microambiente Tumoral/inmunología , Animales , Sitios de Unión/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/biosíntesis , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-6/biosíntesis , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Oxidación-Reducción/efectos de los fármacos , Unión Proteica/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Microambiente Tumoral/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Yao Xue Xue Bao ; 51(2): 294-301, 2016 02.
Artículo en Chino | MEDLINE | ID: mdl-29856584

RESUMEN

Myeloid-derived suppressor cells(MDSC) play critical roles in immune escape of tumor. We hypothesized that elimination of tumor-induced MDSCs might help to block tumor growth. Therefore, we constructed a cholera toxin B based peptide vaccine that targets a MDSC surface marker S100A8. Immunized BALB/c mice with CTB-S100A8 plus aluminum hydroxide induced high titers of anti-S100A8 antibodies and reduced tumor burden significantly in 4T1 mice model. We also found the vaccination led to significant reduction of tumor-induced monocytic MDSC(M-MDSC), with no effect on innate MDSCs, dendritic cell(DC) and macrophage(Mφ), demonstrating that targeting tumor-induced MDSC may be a promising approach in cancer immunotherapy.


Asunto(s)
Calgranulina A/antagonistas & inhibidores , Vacunas contra el Cáncer , Células Mieloides/efectos de los fármacos , Neoplasias/terapia , Vacunas de Subunidad , Animales , Línea Celular Tumoral , Macrófagos , Ratones , Ratones Endogámicos BALB C
12.
J Leukoc Biol ; 97(4): 677-88, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25713087

RESUMEN

MDSCs are potent immunosuppressive cells that are induced during inflammatory responses, as well as by cancers, to evade the anti-tumor immunity. We recently demonstrated that marijuana cannabinoids are potent inducers of MDSCs. In the current study, we investigated the epigenetic mechanisms through which THC, an exogenous cannabinoid, induces MDSCs and compared such MDSCs with the naïve MDSCs found in BM of BL6 (WT) mice. Administration of THC into WT mice caused increased methylation at the promoter region of DNMT3a and DNMT3b in THC-induced MDSCs, which correlated with reduced expression of DNMT3a and DNMT3b. Furthermore, promoter region methylation was decreased at Arg1 and STAT3 in THC-induced MDSCs, and consequently, such MDSCs expressed higher levels of Arg1 and STAT3. In addition, THC-induced MDSCs secreted elevated levels of S100A8, a calcium-binding protein associated with accumulation of MDSCs in cancer models. Neutralization of S100A8 by use of anti-S100A8 (8H150) in vivo reduced the ability of THC to trigger MDSCs. Interestingly, the elevated S100A8 expression also promoted the suppressive function of MDSCs. Together, the current study demonstrates that THC mediates epigenetic changes to promote MDSC differentiation and function and that S100A8 plays a critical role in this process.


Asunto(s)
Calgranulina A/fisiología , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Células Mieloides/inmunología , Regiones Promotoras Genéticas/efectos de los fármacos , Factor de Transcripción STAT3/fisiología , Animales , Arginasa/biosíntesis , Arginasa/genética , Calgranulina A/antagonistas & inhibidores , Calgranulina A/biosíntesis , Calgranulina A/genética , Diferenciación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Dronabinol , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética , Organismos Libres de Patógenos Específicos , Linfocitos T Reguladores/inmunología , ADN Metiltransferasa 3B
13.
Anticancer Res ; 34(4): 1873-84, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24692722

RESUMEN

BACKGROUND/AIM: The proinflammatory protein S100A8, which is expressed in myeloid cells under physiological conditions, is strongly expressed in human cancer tissues. Its role in tumor cell differentiation and tumor progression is largely unclear and virtually unstudied in kidney cancer. In the present study, we investigated whether S100A8 could be a potential anticancer drug target and therapeutic biomarker for kidney cancer, and the underlying molecular mechanisms by exploiting its interaction profile with drugs. MATERIALS AND METHODS: Microarray-based transcriptomics experiments using Affymetrix HuGene 1.0 ST arrays were applied to renal cell carcinoma specimens from Saudi patients for identification of significant genes associated with kidney cancer. In addition, we retrieved selected expression data from the National Center for Biotechnology Information Gene Expression Omnibus database for comparative analysis and confirmation of S100A8 expression. Ingenuity Pathway Analysis (IPA) was used to elucidate significant molecular networks and pathways associated with kidney cancer. The probable polar and non-polar interactions of possible S100A8 inhibitors (aspirin, celecoxib, dexamethasone and diclofenac) were examined by performing molecular docking and binding free energy calculations. Detailed analysis of bound structures and their binding free energies was carried out for S100A8, its known partner (S100A9), and S100A8-S100A9 complex (calprotectin). RESULTS: In our microarray experiments, we identified 1,335 significantly differentially expressed genes, including S100A8, in kidney cancer using a cut-off of p<0.05 and fold-change of 2. Functional analysis of kidney cancer-associated genes showed overexpression of genes involved in cell-cycle progression, DNA repair, cell death, tumor morphology and tissue development. Pathway analysis showed significant disruption of pathways of atherosclerosis signaling, liver X receptor/retinoid X receptor (LXR/RXR) activation, notch signaling, and interleukin-12 (IL-12) signaling. We identified S100A8 as a prospective biomarker for kidney cancer and in silico analysis showed that aspirin, celecoxib, dexamethasone and diclofenac binds to S100A8 and may inhibit downstream signaling in kidney cancer. CONCLUSION: The present study provides an initial overview of differentially expressed genes in kidney cancer of Saudi Arabian patients using whole-transcript, high-density expression arrays. Our analysis suggests distinct transcriptomic signatures, with significantly high levels of S100A8, and underlying molecular mechanisms contributing to kidney cancer progression. Our docking-based findings shed insight into S100A8 protein as an attractive anticancer target for therapeutic intervention in kidney cancer. To our knowledge, this is the first structure-based docking study for the selected protein targets using the chosen ligands.


Asunto(s)
Antineoplásicos/química , Calgranulina A/química , Calgranulina A/genética , Expresión Génica , Neoplasias Renales/genética , Neoplasias Renales/patología , Simulación del Acoplamiento Molecular , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Calgranulina A/antagonistas & inhibidores , Calgranulina A/metabolismo , Análisis por Conglomerados , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Neoplasias Renales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Estadificación de Neoplasias , Unión Proteica , Transducción de Señal , Carga Tumoral
14.
J Clin Invest ; 123(11): 4595-611, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24216507

RESUMEN

Myelodysplastic syndromes (MDS) are age-dependent stem cell malignancies that share biological features of activated adaptive immune response and ineffective hematopoiesis. Here we report that myeloid-derived suppressor cells (MDSC), which are classically linked to immunosuppression, inflammation, and cancer, were markedly expanded in the bone marrow of MDS patients and played a pathogenetic role in the development of ineffective hematopoiesis. These clonally distinct MDSC overproduce hematopoietic suppressive cytokines and function as potent apoptotic effectors targeting autologous hematopoietic progenitors. Using multiple transfected cell models, we found that MDSC expansion is driven by the interaction of the proinflammatory molecule S100A9 with CD33. These 2 proteins formed a functional ligand/receptor pair that recruited components to CD33's immunoreceptor tyrosine-based inhibition motif (ITIM), inducing secretion of the suppressive cytokines IL-10 and TGF-ß by immature myeloid cells. S100A9 transgenic mice displayed bone marrow accumulation of MDSC accompanied by development of progressive multilineage cytopenias and cytological dysplasia. Importantly, early forced maturation of MDSC by either all-trans-retinoic acid treatment or active immunoreceptor tyrosine-based activation motif­bearing (ITAM-bearing) adapter protein (DAP12) interruption of CD33 signaling rescued the hematologic phenotype. These findings indicate that primary bone marrow expansion of MDSC driven by the S100A9/CD33 pathway perturbs hematopoiesis and contributes to the development of MDS.


Asunto(s)
Síndromes Mielodisplásicos/etiología , Células Mieloides/inmunología , Traslado Adoptivo , Animales , Calgranulina A/antagonistas & inhibidores , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Microambiente Celular , Modelos Animales de Enfermedad , Femenino , Antígenos HLA-DR/metabolismo , Hematopoyesis/inmunología , Humanos , Ligandos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Síndromes Mielodisplásicos/inmunología , Síndromes Mielodisplásicos/patología , Células Mieloides/patología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal/inmunología
15.
Leukemia ; 26(6): 1255-65, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22282267

RESUMEN

MLL-rearranged acute lymphoblastic leukemia (ALL) in infants is characterized by a poor clinical outcome and resistance to glucocorticoids (for example, prednisone and dexamethasone). As both the response to prednisolone in vitro and prednisone in vivo are predictive for clinical outcome, understanding and overcoming glucocorticoid resistance remains an essential step towards improving prognosis. Prednisolone-induced apoptosis depends on glucocorticoid-evoked Ca(2+) fluxes from the endoplasmic reticulum towards the mitochondria. Here, we demonstrate that in MLL-rearranged infant ALL, over-expression of S100A8 and S100A9 is associated with failure to induce free-cytosolic Ca(2+) and prednisolone resistance. Furthermore, we demonstrate that enforced expression of S100A8/S100A9 in prednisolone-sensitive MLL-rearranged ALL cells, rapidly leads to prednisolone resistance as a result of S100A8/S100A9 mediated suppression of prednisolone-induced free-cytosolic Ca(2+) levels. In addition, the Src kinase inhibitor PP2 markedly sensitized MLL-rearranged ALL cells otherwise resistant to prednisolone, via downregulation of S100A8 and S100A9, which allowed prednisolone-induced Ca(2+) fluxes to reach the mitochondria and trigger apoptosis. On the basis of this novel mechanism of prednisolone resistance, we propose that developing more specific S100A8/S100A9 inhibitors may well be beneficial for prednisolone-resistant MLL-rearranged infant ALL patients.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Resistencia a Antineoplásicos , Reordenamiento Génico , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Prednisolona/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Calgranulina A/antagonistas & inhibidores , Calgranulina A/genética , Calgranulina B/administración & dosificación , Calgranulina B/genética , Citometría de Flujo , Estudios de Seguimiento , Glucocorticoides/farmacología , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Recién Nacido , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Prednisona/farmacología , Pronóstico , Pirimidinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Tasa de Supervivencia , Familia-src Quinasas/antagonistas & inhibidores
16.
Oncol Rep ; 27(3): 813-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22089965

RESUMEN

Dendrobium nobile is widely used as an analgesic, an antipyretic, and a tonic to nourish the stomach in traditional medicine. Mounting evidence suggests an antitumor activity of denbinobin, a major phenanthrene isolated from stems of Dendrobium nobile. The present study aimed to investigate the inhibitory effect of denbinobin on the invasive ability of human cancer cells. The cytotoxicity of denbonobin was examined in several human cancer cell lines including SK-Hep-1 hepato-carcinoma cells, SNU-484 gastric cancer cells, and HeLa cervix cancer cells. Because SNU-484 cells showed the lowest IC50 value, we examined the effect of denbinobin on the invasive ability of SNU-484 cells. The present study revealed, for the first time, that denbinobin inhibits the invasive phenotype of SNU-484 human gastric cancer cells in a dose-dependent manner. Expressions of matrix metalloproteinase (MMP)-2 and MMP-9 were significantly decreased by denbinobin, suggesting that MMP-2/-9 may be responsible for the anti-invasive activity of denbinobin. We also provide evidence that denbinobin induces apoptosis through down-regulation of Bcl-2 and an up-regulation of Bax. Taken together, this study demonstrates that denbinobin inhibits invasion and induces apoptosis in highly invasive SNU-484 human gastric cancer cells. Given that gastric cancer has been estimated to be one of the most common causes of cancer-related death among Asians and the major cause of death from gastric cancer is the metastatic spread of the disease, our findings may provide useful information regarding the application of denbinobin as a chemopreventive agent that could prevent or alleviate metastatic gastric cancer.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Fenantrenos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Calgranulina A/antagonistas & inhibidores , Calgranulina A/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioprevención/métodos , Dendrobium/química , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Células HeLa , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Invasividad Neoplásica , Fenotipo , Preparaciones de Plantas/farmacología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
17.
J Immunol ; 183(7): 4583-92, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19752232

RESUMEN

Finegoldia magna is an anaerobic bacterial species that is part of the normal human flora on all nonsterile body surfaces, but it is also a significant opportunistic pathogen causing a wide range of infections. Some isolates of F. magna that are more frequently associated with clinical infection express protein L, a surface protein containing multiple homologous domains (B1-B5) that bind Igs through interactions with Ig L chains. The present study shows that the N-terminal A domain of protein L binds S100A8/A9, antibacterial proteins present in large amounts in the cytoplasm of neutrophils, but also extracellularly in tissues during inflammation. As a result, protein L-expressing F. magna are protected against killing by S100A8/A9. Igs and S100A8/A9 were found to interact independently with protein L, demonstrating that this bacterial surface protein is capable of manipulating both adaptive and innate immune defense mechanisms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/fisiología , Calgranulina A/antagonistas & inhibidores , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/microbiología , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/biosíntesis , Bacteriólisis/inmunología , Actividad Bactericida de la Sangre/inmunología , Calgranulina A/fisiología , Calgranulina B/fisiología , Proteínas de Unión al ADN/biosíntesis , Humanos , Neutrófilos/metabolismo , Peptococcus/crecimiento & desarrollo , Peptococcus/inmunología , Peptococcus/metabolismo , Unión Proteica/inmunología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/metabolismo , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/metabolismo
18.
J Immunol ; 180(5): 3366-74, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18292562

RESUMEN

We investigated the roles of the potent, chemotactic antimicrobial proteins S100A8, S100A9, and S100A8/A9 in leukocyte migration in a model of streptococcal pneumonia. We first observed differential secretion of S100A8, S100A9, and S100A8/A9 that preceded neutrophil recruitment. This is partially explained by the expression of S100A8 and S100A9 proteins by pneumocytes in the early phase of Streptococcus pneumoniae infection. Pretreatment of mice with anti-S100A8 and anti-S100A9 Abs, alone or in combination had no effect on bacterial load or mice survival, but caused neutrophil and macrophage recruitment to the alveoli to diminish by 70 and 80%, respectively, without modifying leukocyte blood count, transendothelial migration or neutrophil sequestration in the lung vasculature. These decreases were also associated with a 68% increase of phagocyte accumulation in lung tissue and increased expression of the chemokines CXCL1, CXCL2, and CCL2 in lung tissues and bronchoalveolar lavages. These results show that S100A8 and S100A9 play an important role in leukocyte migration and strongly suggest their involvement in the transepithelial migration of macrophages and neutrophils. They also indicate the importance of antimicrobial proteins, as opposed to classical chemotactic factors such as chemokines, in regulating innate immune responses in the lung.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/antagonistas & inhibidores , Péptidos Catiónicos Antimicrobianos/metabolismo , Calgranulina A/antagonistas & inhibidores , Calgranulina B/inmunología , Inhibición de Migración Celular/inmunología , Fagocitos/inmunología , Neumonía Bacteriana/inmunología , Alveolos Pulmonares/inmunología , Infecciones Estreptocócicas/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Calgranulina A/inmunología , Calgranulina A/fisiología , Calgranulina B/fisiología , Modelos Animales de Enfermedad , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/ultraestructura , Ratones , Monocitos/inmunología , Monocitos/patología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Fagocitos/patología , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/microbiología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Streptococcus pneumoniae/inmunología , Factores de Tiempo
19.
J Immunol ; 171(5): 2602-9, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12928412

RESUMEN

Recently, proinflammatory activities had been described for S100A8 and S100A9, two proteins found at inflammatory sites and within the neutrophil cytoplasm. In this study, we investigated the role of these proteins in neutrophil migration in vivo in response to LPS. LPS was injected into the murine air pouch, which led to the release of S100A8, S100A9, and S100A8/A9 in the pouch exudates that preceded accumulation of neutrophils. Passive immunization against S100A8 and S100A9 led to a 52% inhibition of neutrophil migration in response to LPS at 3 h postinjection. Injection of LPS was also associated with an increase in peripheral blood neutrophils and the presence in serum of S100A9 and S100A8/A9. Intravenous injection of S100A8, S100A9, or S100A8/A9 augmented the number of circulating neutrophils and diminished the number of neutrophils in the bone marrow, demonstrating that S100A8 and S100A9 induced the mobilization of neutrophils from the bone marrow to the blood. Finally, passive immunization with anti-S100A9 inhibited the neutrophilia associated with LPS injection in the air pouch. These results suggest that S100A8 and S100A9 play a role in the inflammatory response to LPS by inducing the release of neutrophils from the bone marrow and directing their migration to the inflammatory site.


Asunto(s)
Calgranulina A/antagonistas & inhibidores , Calgranulina A/fisiología , Calgranulina B/fisiología , Inhibición de Migración Celular , Lipopolisacáridos/administración & dosificación , Neutrófilos/citología , Neutrófilos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Calgranulina A/inmunología , Calgranulina A/metabolismo , Calgranulina B/inmunología , Calgranulina B/metabolismo , Agregación Celular/inmunología , Dimerización , Modelos Animales de Enfermedad , Espacio Extracelular/metabolismo , Inmunoglobulina G/administración & dosificación , Inyecciones Intravenosas , Inyecciones Subcutáneas , Leucocitosis/inmunología , Leucocitosis/metabolismo , Leucocitosis/patología , Leucocitosis/prevención & control , Ratones , Neutrófilos/patología , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA