Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Crit Care ; 27(1): 374, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773186

RESUMEN

BACKGROUND AND AIMS: The triggering factors of sepsis-induced myocardial dysfunction (SIMD) are poorly understood and are not addressed by current treatments. S100A8/A9 is a pro-inflammatory alarmin abundantly secreted by activated neutrophils during infection and inflammation. We investigated the efficacy of S100A8/A9 blockade as a potential new treatment in SIMD. METHODS: The relationship between plasma S100A8/A9 and cardiac dysfunction was assessed in a cohort of 62 patients with severe sepsis admitted to the intensive care unit of Linköping University Hospital, Sweden. We used S100A8/A9 blockade with the small-molecule inhibitor ABR-238901 and S100A9-/- mice for therapeutic and mechanistic studies on endotoxemia-induced cardiac dysfunction in mice. RESULTS: In sepsis patients, elevated plasma S100A8/A9 was associated with left-ventricular (LV) systolic dysfunction and increased SOFA score. In wild-type mice, 5 mg/kg of bacterial lipopolysaccharide (LPS) induced rapid plasma S100A8/A9 increase and acute LV dysfunction. Two ABR-238901 doses (30 mg/kg) administered intraperitoneally with a 6 h interval, starting directly after LPS or at a later time-point when LV dysfunction is fully established, efficiently prevented and reversed the phenotype, respectively. In contrast, dexamethasone did not improve cardiac function compared to PBS-treated endotoxemic controls. S100A8/A9 inhibition potently reduced systemic levels of inflammatory mediators, prevented upregulation of inflammatory genes and restored mitochondrial function in the myocardium. The S100A9-/- mice were protected against LPS-induced LV dysfunction to an extent comparable with pharmacologic S100A8/A9 blockade. The ABR-238901 treatment did not induce an additional improvement of LV function in the S100A9-/- mice, confirming target specificity. CONCLUSION: Elevated S100A8/A9 is associated with the development of LV dysfunction in severe sepsis patients and in a mouse model of endotoxemia. Pharmacological blockade of S100A8/A9 with ABR-238901 has potent anti-inflammatory effects, mitigates myocardial dysfunction and might represent a novel therapeutic strategy for patients with severe sepsis.


Asunto(s)
Endotoxemia , Cardiopatías , Disfunción Ventricular Izquierda , Humanos , Ratones , Animales , Endotoxemia/complicaciones , Endotoxemia/tratamiento farmacológico , Lipopolisacáridos , Calgranulina A/fisiología , Calgranulina B/genética , Miocardio , Inflamación/tratamiento farmacológico
2.
Int J Mol Sci ; 22(20)2021 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-34681835

RESUMEN

Paracrine factors of human mesenchymal stem cells (hMSCs) have the potential of preventing adverse cardiac remodeling after myocardial infarction (MI). S100A8 and S100A9 are calcium-binding proteins playing essential roles in the regulation of inflammation and fibrous tissue formation, and they might modulate the paracrine effect of hMSCs. We isolated human amniotic mesenchymal stem cells (hAMSCs) and examined the changes in the expression level of regulatory genes of inflammation and fibrosis after hAMSCs were treated with S100A8/A9. The anti-inflammatory and anti-fibrotic effects of hAMSCs pretreated with S100A8/A9 were shown to be superior to those of hAMSCs without S100A8/A9 pretreatment in the cardiomyocyte hypoxia/reoxygenation experiment. We established a murine myocardial ischemia/reperfusion model to compare the therapeutic effects of the conditioned medium of hAMSCs with or without S100A8/A9 pretreatment. We found the hearts administered with a conditioned medium of hAMSCs with S100A8/A9 pretreatment had better left ventricular systolic function on day 7, 14, and 28 after MI. These results suggest S100A8/A9 enhances the paracrine therapeutic effects of hAMSCs in aspects of anti-inflammation, anti-fibrosis, and cardiac function preservation after MI.


Asunto(s)
Calgranulina A/fisiología , Calgranulina B/fisiología , Inmunomodulación , Células Madre Mesenquimatosas/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Proteínas de Unión al Calcio/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Regulación de la Expresión Génica , Humanos , Agentes Inmunomoduladores/farmacología , Inflamación/metabolismo , Isquemia/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo
3.
FASEB J ; 35(9): e21798, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34339064

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic threatens human species with mortality rate of roughly 2%. We can hardly predict the time of herd immunity against and end of COVID-19 with or without success of vaccine. One way to overcome the situation is to define what delineates disease severity and serves as a molecular target. The most successful analogy is found in BCR-ABL in chronic myeloid leukemia, which is the golden biomarker, and simultaneously, the most effective molecular target. We hypothesize that S100 calcium-binding protein A8 (S100A8) is one such molecule. The underlying evidence includes accumulating clinical information that S100A8 is upregulated in severe forms of COVID-19, pathological similarities of the affected lungs between COVID-19 and S100A8-induced acute respiratory distress syndrome (ARDS) model, homeostatic inflammation theory in which S100A8 is an endogenous ligand for endotoxin sensor Toll-like receptor 4/Myeloid differentiation protein-2 (TLR4/MD-2) and mediates hyper-inflammation even after elimination of endotoxin-producing extrinsic pathogens, analogous findings between COVID-19-associated ARDS and pre-metastatic lungs such as S100A8 upregulation, pulmonary recruitment of myeloid cells, increased vascular permeability, and activation coagulation cascade. A successful treatment in an animal COVID-19 model is given with a reagent capable of abrogating interaction between S100A8/S100A9 and TLR4. In this paper, we try to verify our hypothesis that S100A8 governs COVID-19-associated ARDS.


Asunto(s)
COVID-19/complicaciones , Calgranulina A/fisiología , Síndrome de Liberación de Citoquinas/etiología , Inflamación/etiología , Pandemias , Síndrome de Dificultad Respiratoria/etiología , SARS-CoV-2/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/fisiología , Animales , Antivirales/farmacología , COVID-19/genética , COVID-19/patología , Calgranulina A/sangre , Calgranulina A/genética , Quimiocina CXCL11/sangre , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/patología , Disacáridos/farmacología , Disacáridos/uso terapéutico , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Inflamación/genética , Inflamación/patología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Antígeno 96 de los Linfocitos/fisiología , Macaca mulatta , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/metabolismo , Especificidad de la Especie , Fosfatos de Azúcar/farmacología , Fosfatos de Azúcar/uso terapéutico , Receptor Toll-Like 4/fisiología , Regulación hacia Arriba , Internalización del Virus
4.
Front Immunol ; 12: 643144, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717200

RESUMEN

Glucose-dependent insulinotropic polypeptide (GIP) communicates information on energy availability from the gut to peripheral tissues. Disruption of its signaling in myeloid immune cells during high-fat diet (HFD)-induced obesity impairs energy homeostasis due to the unrestrained metabolically deleterious actions of S100A8/A9 alarmin. White adipose tissue (WAT) type 2 immune cell networks are important for maintaining metabolic and energy homeostasis and limiting obesity-induced inflammation. Nevertheless, the consequences of losing immune cell GIP receptor (GIPR) signaling on type 2 immunity in WAT remains unknown. Bone marrow (BM) chimerism was used to generate mice with GIPR (Gipr-/- BM) and GIPR/S100A8/A9 (Gipr-/- /S100a9-/- BM) deletion in immune cells. These mice were subjected to short (5 weeks) and progressive (14 weeks) HFD regimens. GIPR-deficiency was also targeted to myeloid cells by crossing Giprfl/fl mice and Lyz2cre/+ mice (LysMΔGipr ). Under both short and progressive HFD regimens, Gipr-/- BM mice exhibited altered expression of key type 2 immune cytokines in the epididymal visceral WAT (epiWAT), but not in subcutaneous inguinal WAT. This was further linked to declined representation of type 2 immune cells in epiWAT, such as group 2 innate lymphoid cells (ILC2), eosinophils, and FOXP3+ regulatory T cells (Tregs). Co-deletion of S100A8/A9 in Gipr-/- immune cells reversed the impairment of type 2 cytokine expression in epiWAT, suggesting a mechanistic role for this alarmin in type 2 immune suppression. LysMΔGipr mice on HFD also displayed altered expression of type 2 immune mediators, highlighting that GIPR-deficiency in myeloid immune cells is responsible for the impairment of type 2 immune networks. Finally, abrogated GIPR signaling in immune cells also affected adipocyte fraction cells, inducing their increased production of the beiging interfering cytokine IL-10 and stress- related type 2 cytokine IL-13. Collectively, these findings attribute an important role for GIPR in myeloid immune cells in supporting WAT type 2 immunity.


Asunto(s)
Tejido Adiposo Blanco/inmunología , Linfocitos/inmunología , Obesidad/inmunología , Receptores de la Hormona Gastrointestinal/fisiología , Tejido Adiposo Blanco/metabolismo , Animales , Calgranulina A/fisiología , Calgranulina B/fisiología , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Transducción de Señal/fisiología , Termogénesis
5.
Oncol Rep ; 45(1): 151-158, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33169801

RESUMEN

B­cell lymphomas (BCLs) are malignant lymphoid tumours originating from the malignant proliferation and transformation of mature lymphocytes at various stages of differentiation and clonal expansion of the lymphatic and circulatory systems. Efforts to control or even eradicate BCLs are frequently hampered by the development of drug resistance. Autophagy is an evolutionarily conserved biological process of the energy metabolism. By degrading intracellular organelles and proteins, autophagy provides cells with biochemical reaction substrates for the maintenance of homeostasis under nutrient deprivation or other stressful conditions. Accumulating evidence indicates that autophagy plays an important role in chemotherapy resistance. S100A8 is an important member of the calcium­binding protein family that plays an important role in regulating tumour resistance to chemotherapy, while the specific molecular regulatory mechanisms remain unclear. In the present study, by employing three BCL cell lines (Daudi, SUDHL­4 and JeKo­1), it was demonstrated that BCL cells with a strong drug resistance also exhibited active autophagy. In addition, S100A8 was found to be crucial for regulating drug resistance and promoting autophagy in BCL cells. Interference of S100A8 significantly downregulated Bcl­2/adenovirus E1B 19­kDa protein­interacting protein 3 located in the mitochondria and endoplasmic reticulum to further inhibit autophagy. In addition, S100A8 interference markedly inhibited the formation of the BECN1­PI3KC3 complex and promoted B­cell lymphoma 2 expression, which collectively inhibited autophagy.


Asunto(s)
Autofagia/fisiología , Calgranulina A/fisiología , Linfoma de Células B/tratamiento farmacológico , Autofagia/efectos de los fármacos , Beclina-1/análisis , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasas Clase III/análisis , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Humanos , Linfoma de Células B/patología , Proteínas de la Membrana/análisis , Proteínas Proto-Oncogénicas/análisis , Vincristina/farmacología
6.
Cell Rep ; 33(13): 108571, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33378668

RESUMEN

Here, we report that functional heterogeneity of macrophages in cancer could be determined by the nature of their precursors: monocytes (Mons) and monocytic myeloid-derived suppressor cells (M-MDSCs). Macrophages that are differentiated from M-MDSCs, but not from Mons, are immune suppressive, with a genomic profile matching that of M-MDSCs. Immune-suppressive activity of M-MDSC-derived macrophages is dependent on the persistent expression of S100A9 protein in these cells. S100A9 also promotes M2 polarization of macrophages. Tissue-resident- and Mon-derived macrophages lack expression of this protein. S100A9-dependent immune-suppressive activity of macrophages involves transcription factor C/EBPß. The presence of S100A9-positive macrophages in tumor tissues is associated with shorter survival in patients with head and neck cancer and poor response to PD-1 antibody treatment in patients with metastatic melanoma. Thus, this study reveals the pathway of the development of immune-suppressive macrophages and suggests an approach to their selective targeting.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Calgranulina A/fisiología , Calgranulina B/fisiología , Terapia de Inmunosupresión , Macrófagos/metabolismo , Monocitos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/inmunología , Microambiente Tumoral
7.
Cell Death Dis ; 11(8): 650, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32811814

RESUMEN

Metastasis is the main cause of failure of cancer treatment. Metastatic colonization is regarded the most rate-limiting step of metastasis and is subjected to regulation by a plethora of biological factors and processes. On one hand, regulation of metastatic colonization by autophagy appears to be stage- and context-dependent, whereas mechanistic characterization remains elusive. On the other hand, interactions between the tumor cells and their microenvironment in metastasis have long been appreciated, whether the secretome of tumor cells can effectively reshape the tumor microenvironment has not been elucidated mechanistically. In the present study, we have identified "SEC23A-S1008-BECLIN1-autophagy axis" in the autophagic regulation of metastatic colonization step, a mechanism that tumor cells can exploit autophagy to exert self-restrain for clonogenic proliferation before the favorable tumor microenvironment is established. Specifically, we employed a paired lung-derived oligometastatic cell line (OL) and the homologous polymetastatic cell line (POL) from human melanoma cell line M14 that differ in colonization efficiency. We show that S100A8 transported by SEC23A inhibits metastatic colonization via autocrine activation of autophagy. Furthermore, we verified the clinical relevance of our experimental findings by bioinformatics analysis of the expression of Sec23a and S100A8 and the clinical-pathological associations. We demonstrate that higher Sec23a and Atg5 expression levels appear to be protective factors and favorable diagnostic (TNM staging) and prognostic (overall survival) markers for skin cutaneous melanoma (SKCM) and colon adenocarcinoma (COAD) patients. And we confirm the bioinformatics analysis results with SKCM biopsy samples.


Asunto(s)
Calgranulina A/metabolismo , Metástasis de la Neoplasia/patología , Proteínas de Transporte Vesicular/metabolismo , Animales , Comunicación Autocrina/fisiología , Autofagia/fisiología , Beclina-1/metabolismo , Calgranulina A/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Humanos , Masculino , Melanoma/patología , Ratones SCID , Pronóstico , ARN Interferente Pequeño/metabolismo , Neoplasias Cutáneas/patología , Microambiente Tumoral/fisiología , Proteínas de Transporte Vesicular/fisiología
8.
Front Immunol ; 11: 688, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425933

RESUMEN

The restricted capacity of newborn infants to mount inflammatory responses toward microbial challenges has traditionally been linked to the high risk of septic diseases during the neonatal period. In recent years, substantial evidence has been provided that this characteristic of the neonatal immune system is actually a meaningful physiologic state that is based on specific transiently active cellular and molecular mechanisms and required for a favorable course of postnatal immune adaptation. The identification of physiologically high amounts of S100-alarmins in neonates has been one of the crucial pieces in the puzzle that contributed to the change of concept. In this context, innate immune immaturity could be redefined and assigned to the epigenetic silence of adult-like cell-autonomous regulation at the beginning of life. S100-alarmins represent an alternative age-specific mechanism of immune regulation that protects neonates from hyperinflammatory immune responses. Here, we summarize how infants are provided with S100-alarmins and why these allow an uneventful clash between the innate immune system and the extrauterine world. The mode of action of S100-alarmins is highlighted including their tuning functions at multiple levels for establishing a state of homeostasis with the environment in the newborn individual.


Asunto(s)
Alarminas/fisiología , Calgranulina A/fisiología , Calgranulina B/fisiología , Inmunidad Innata , Recién Nacido/inmunología , Adaptación Fisiológica , Humanos , Transducción de Señal/fisiología
9.
Pathog Dis ; 78(5)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31116394

RESUMEN

The majority of chronic wounds are associated with bacterial biofilms recalcitrant to antibiotics and host responses. Immunomodulatory S100A8/A9 is suppressed in Pseudomonas aeruginosa biofilm infected wounds. We aimed at investigating a possible additive effect between S100A8/A9 and ciprofloxacin against biofilms. MATERIALS/METHODS: Thirty-two mice were injected with alginate-embedded P. aeruginosa following a third-degree burn. The mice were randomized into four groups receiving combination ciprofloxacin and S100A8/A9 or monotherapy ciprofloxacin, S100A8/A9 or a placebo and evaluated by host responses and quantitative bacteriology in wounds. In addition, in vitro checkerboard analysis was performed, with P. aeruginosa and ascending S100A8/A9 and ciprofloxacin concentrations. RESULTS: S100A8/A9 augmented the effect of ciprofloxacin in vivo by lowering the bacterial quantity compared to the placebo arm and the two monointervention groups (P < 0.0001). S100A8 and 100A9 were increased in the double-treated group as compared to the monointervention groups (P = 0.032, P = 0.0023). Tissue inhibitor of metalloproteinases-1 and keratinocyte\chemokine chemoattractant-1 were increased in the double-intervention group compared to the S100A8/A9 group (P = 0.050, P = 0.050). No in vitro synergism was detected. CONCLUSION: The observed ciprofloxacin-augmenting effect of S100A8/A9 in vivo was not confirmed by checkerboard analysis, indicating dependence on host cells for the S100A8/A9 effect. S100A8/A9 and ciprofloxacin is a promising therapy for optimizing chronic wound treatment.


Asunto(s)
Biopelículas/efectos de los fármacos , Calgranulina A/fisiología , Ciprofloxacina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Infección de Heridas/inmunología , Infección de Heridas/microbiología , Animales , Antibacterianos/farmacología , Enfermedad Crónica , Recuento de Colonia Microbiana , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología
10.
J Leukoc Biol ; 106(1): 35-43, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31091351

RESUMEN

Intravascular hemolysis, in addition to reducing red cell counts, incurs extensive vascular inflammation and oxidative stress. One product of hemolysis, heme, is a potent danger associated molecular pattern (DAMP), activating leukocytes and inducing cytokine expression and processing, among other pro-inflammatory effects. We explored pathways by which heme-induced inflammation may be amplified under sterile conditions. Incubation of human Mϕs, differentiated from CD14+ cells, with heme induced time- and concentration-dependent gene and protein expression of S100A8, a myeloid cell-derived alarmin. Human Mϕ stimulation with recombinant S100A8, in turn, induced robust pro-IL-1ß expression that was dependent upon NF-κB activation, gene transcription, and partially dependent upon TLR4-mediated signaling. Moreover, heme itself stimulated significant Mϕ pro-IL-1ß gene and protein expression via an S100A8-mediated mechanism and greatly amplified S100A8-driven NLRP3 inflammasome-mediated IL-1ß secretion. In vivo, induction of acute intravascular hemolysis in mice induced a rapid elevation of plasma S100A8 that could be abolished by hemopexin, a heme scavenger. Finally, plasma S100A8 levels were found to be significantly elevated in patients with the inherited hemolytic anemia, sickle cell anemia, when compared with levels in healthy individuals. In conclusion, we demonstrate that hemolytic processes are associated with S100A8 generation and that some of the inflammatory effects of heme may be amplified by autocrine S100A8 production. Findings suggest a mechanism by which hemolytic inflammation could be propagated via leukocyte priming by endogenous proteins, even in sterile inflammatory environments such as those that occur in the hemolytic diseases. S100A8 may represent a therapeutic target for reducing inflammation in hemolytic disorders.


Asunto(s)
Calgranulina A/fisiología , Hemo/farmacología , Hemólisis/inmunología , Inflamación/inmunología , Macrófagos/efectos de los fármacos , Adulto , Animales , Femenino , Humanos , Interleucina-1beta/fisiología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , Receptor Toll-Like 4/fisiología
11.
Biochem Pharmacol ; 165: 170-180, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30862503

RESUMEN

For more than two centuries now, rheumatoid arthritis (RA) is under investigation intending to discover successful treatment. Despite decades of scientific advances, RA is still representing a challenge for contemporary medicine. Current drug therapies allow to improve significantly the quality of life of RA patients; however, they are still insufficient to reverse tissue injury and are often generating side-effects. The difficulty arises from the considerable fluctuation of the clinical course of RA among patients, making the predictive prognosis difficult. More and more studies underline the profound influence of the neutrophil multifaceted functions in the pathogenesis of RA. This renewed interest in the complexity of neutrophil functions in RA offers new exciting opportunities for valuable therapeutic targets as well as for safe and well-tolerated RA treatments. In this review, we aim to update the recent findings on the multiple facets of neutrophils in RA, in particular their impact in promoting the RA-based inflammation through the release of the cytokine-like S100A8/A9 protein complex, as well as the importance of NETosis in the disease progression and development. Furthermore, we delve into the complex question of neutrophil heterogeneity and plasticity and discuss the emerging role of miRNAs and epigenetic markers influencing the inflammatory response of neutrophils in RA and how they could constitute the starting point for novel attractive targets in RA therapy.


Asunto(s)
Artritis Reumatoide/etiología , Neutrófilos/fisiología , Animales , Artritis Reumatoide/inmunología , Calgranulina A/fisiología , Calgranulina B/fisiología , Epigénesis Genética , Trampas Extracelulares/fisiología , Humanos , MicroARNs/fisiología , NADPH Oxidasas/fisiología , Especies Reactivas de Oxígeno/metabolismo
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(7): 627-631, 2018 Jul.
Artículo en Chino | MEDLINE | ID: mdl-30381127

RESUMEN

Objective To investigate the effects of myeloid-related protein 8 (MRP8), MRP14 and MRP8/14 heterodimer on the phenotypic maturation of mice bone marrow-derived dendritic cells (BMDCs). Methods BMDCs were cultured and purified in vitro and divided into control group (equal volume of PBS), MRP14 (1 µg/mL) treatment group, MRP8 (1 µg/mL) treatment group and MRP8/14 (1 µg/mL) treatment group. Flow cytometry was used to detect the expression of costimulatory molecules, such as CD40, CD80, CD86 and major histocompatibility complex II (MHC II ) on the surface of BMDCs after stimulation. Results MRP14, MRP8 and MRP8/14 promoted the expression of CD40, CD80 and CD86, while MRP14 and MRP8 promoted the expression of MHC II on the surface of BMDCs. Moreover, the ability to promote the expression of CD80 and CD86 is stronger in MRP14 and MRP8/14 than MRP8. Conclusion MRP14, MRP8 and MRP8/14 promote the phenotypic maturation of BMDCs by increasing the expression of costimulatory molecules, and MRP14, MRP8 and MRP8/14 also differ in their ability to promote BMDCs to expresse various costimulatory molecules.


Asunto(s)
Calgranulina A/fisiología , Calgranulina B/fisiología , Células Dendríticas/citología , Animales , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Células de la Médula Ósea , Antígenos CD40/metabolismo , Células Cultivadas , Antígenos de Histocompatibilidad Clase II/metabolismo , Ratones
13.
Brain Res Bull ; 143: 234-245, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30266587

RESUMEN

S100A8/A9, a heterodimer complex composed of calcium-binding proteins S100A8 and S100A9, is significantly increased in the serum of multiple sclerosis (MS) patients. Relevant reports have revealed that MS pathology is commonly associated with the activation of microglial cells and the damage of oligodendrocyte precursor cells (OPCs). Moreover, microglia activation following stimulation increases the expression of pro-inflammatory cytokines, such as interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α), which further exacerbate the damage to OPCs. In this study, we were the first to confirm that S100A8/A9 treatment induced the activation, proliferation and migration of the murine microglia cell line BV-2; moreover, this treatment caused the cells to switch from an anti-inflammatory activated (M2) phenotype to a pro-inflammatory activated (M1) phenotype. Meanwhile, the level of the phosphorylated nuclear factor-κB (p-NF-κB) P65 protein was remarkably elevated, and the production of pro-inflammatory factors (IL-1ß, TNF-α, MMP-9) and chemokines (CCL2, CCL3, CXCL10) was also increased in the S100A8/A9-treated BV-2 microglial cells. Inhibition of NF-κB P65 phosphorylation reversed the effects of S100A8/A9 on the production of pro-inflammatory factors and chemokines. We also explored the effects of S100A8/A9 and S100A8/A9-activated BV-2 microglial cells on the viability of OPCs. The results showed that both the S100A8/A9 complex and the conditioned medium (CM) of the S100A8/A9-activated BV-2 microglial cells resulted in OPC apoptosis, which was more pronounced in the case of the CM treatment. However, OPC apoptosis in the CM group was obviously decreased through the inhibition of NF-κB p65 phosphorylation. This study indicates that S100A8/A9 induces the activation of BV-2 microglial cells and promotes the production of pro-inflammatory factors by activating the NF-κB signaling pathway, which further exacerbates OPC damage.


Asunto(s)
Calgranulina A/fisiología , Calgranulina B/fisiología , Microglía/fisiología , Animales , Apoptosis , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Citocinas/metabolismo , Femenino , Inflamación , Interleucina-1beta/metabolismo , Activación de Macrófagos , Macrófagos/patología , Masculino , FN-kappa B/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/metabolismo , Fosforilación , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
14.
Eur Rev Med Pharmacol Sci ; 22(12): 3925-3935, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29949169

RESUMEN

OBJECTIVE: Inflammation and extracellular matrix degradation play a role in cardiac rupture (CR) after myocardial infarction (MI). It has been found that the expression of inflammatory cytokine S100A8/A9 was elevated in acute MI patients, whereas its impact in CR after infarction remains unclear. PATIENTS AND METHODS: Samples from cardiac tissue and peripheral blood of patients with CR after MI, MI, patients without CR, and healthy control (cardiotrauma) were collected to test the expressions of S100A8/A9, p-p65, and MMP-9. Co-culture system for HCF cells and macrophages were established to identify the impact of hypoxia-ischemia on the expressions of S100A8/A9 and TNFα. S100A9 and/or TNFα blocking agent were applied to examine the effect on macrophages migration, expressions of S100A8, S100A9, and TNFα. Western blot was adopted to determine levels of p-p65 and MMP-9 protein after the inhibition of S100A9 and/or TNFα. RESULTS: Compared with healthy control and non-CR patients, serum S100A8/A9 and MMP-9 levels were elevated in cardiac tissues of CR patients, while S100A8/A9, p-p65, and MMP-9 were also overexpressed. Hypoxia-ischemia significantly caused the increasing levels of S100A8/A9 and TNFα in macrophages (p < 0.05). The blockade of S100A9 and/or TNFα suppressed the activation and migration of macrophages. The inhibition of S100A9 expression also decreased the secretion of TNFα in macrophages, while the suppression of TNFα showed no significant impact on S100A8 and S100A9 levels. Downregulation of TNFα or NF-κB markedly declined p-p65 and MMP-9 protein levels in HCF cells from co-culture system or single culture, whereas the blockade of S100A9 only reduced their expressions in co-cultured HCF cells. CONCLUSIONS: The level of S100A8/A9 was upregulated in MI patients with CR. S100A8/A9 induced the activation of NF-κB and expression of MMP-9 protein in HCF cells through facilitating secretion of TNFα from macrophages, which may play a role in triggering extracellular matrix degradation and CR.


Asunto(s)
Calgranulina A/fisiología , Calgranulina B/fisiología , Rotura Cardíaca/etiología , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Infarto del Miocardio/complicaciones , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular Tumoral , Fibroblastos/metabolismo , Rotura Cardíaca/metabolismo , Humanos , Factor de Transcripción ReIA/metabolismo
15.
J Clin Invest ; 127(6): 2133-2147, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28504650

RESUMEN

Platelets play a critical role in atherogenesis and thrombosis-mediated myocardial ischemia, processes that are accelerated in diabetes. Whether hyperglycemia promotes platelet production and whether enhanced platelet production contributes to enhanced atherothrombosis remains unknown. Here we found that in response to hyperglycemia, neutrophil-derived S100 calcium-binding proteins A8/A9 (S100A8/A9) interact with the receptor for advanced glycation end products (RAGE) on hepatic Kupffer cells, resulting in increased production of IL-6, a pleiotropic cytokine that is implicated in inflammatory thrombocytosis. IL-6 acts on hepatocytes to enhance the production of thrombopoietin, which in turn interacts with its cognate receptor c-MPL on megakaryocytes and bone marrow progenitor cells to promote their expansion and proliferation, resulting in reticulated thrombocytosis. Lowering blood glucose using a sodium-glucose cotransporter 2 inhibitor (dapagliflozin), depleting neutrophils or Kupffer cells, or inhibiting S100A8/A9 binding to RAGE (using paquinimod), all reduced diabetes-induced thrombocytosis. Inhibiting S100A8/A9 also decreased atherogenesis in diabetic mice. Finally, we found that patients with type 2 diabetes have reticulated thrombocytosis that correlates with glycated hemoglobin as well as increased plasma S100A8/A9 levels. These studies provide insights into the mechanisms that regulate platelet production and may aid in the development of strategies to improve on current antiplatelet therapies and to reduce cardiovascular disease risk in diabetes.


Asunto(s)
Aterosclerosis/inmunología , Calgranulina A/fisiología , Calgranulina B/fisiología , Diabetes Mellitus Experimental/inmunología , Neutrófilos/metabolismo , Trombocitosis/inmunología , Animales , Aterosclerosis/metabolismo , Plaquetas/fisiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Trombocitosis/metabolismo
16.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 42(4): 365-373, 2017 Apr 28.
Artículo en Chino | MEDLINE | ID: mdl-28490692

RESUMEN

OBJECTIVE: To identify the interacting proteins with S100A8 or S100A9 in HEK293 cell line by flag-tag affinity purification and liquid chromatography mass spectrometry/mass spectrometry (LC-MS/MS).
 Methods: The p3×Flag-CMV-S100A8 and p3×Flag-CMV-S100A9 expression vectors were constructed by inserting S100A8 or S100A9 coding sequence. The recombinant plasmids were then transfected into HEK293 cells. Affinity purification and LC-MS/MS were applied to identify the proteins interacting with S100A8 or S100A9. Bioinformatics analysis was used to seek the gene ontology of the interacting proteins. Co-immunoprecipitation (Co-IP) was applied to confirm the proteins interacted with S100A8 or S100A9.
 Results: Fourteen proteins including pyruvate kinase, muscle (PKM), nucleophosmin (NPM1) and eukaryotic translation initiation factor 5A (EIF5A), which potentially interacted with S100A8, were successfully identified by Flag-tag affinity purification followed by LC-MS/MS analysis. Six proteins, such as tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (14-3-3ε) and PKM, which potentially interacted with S100A9, were successfully identified. Gene ontology analysis of the identified proteins suggested that proteins interacted with S100A8 or S100A9 were involved in several biological pathways, including canonical glycolysis, positive regulation of NF-κB transcription factor activity, negative regulation of apoptotic process, cell-cell adhesion, etc. Co-IP experiment confirmed that PKM2 can interact with both S100A8 and S100A9, and 14-3-3ε can interact with S100A8.
 Conclusion: PKM2 is identified to interact with both S100A8 and S100A9, while 14-3-3ε can interact with S100A9. These results may provide a new clue for the role of S100A8 or S100A9 in the progression of colitis-associated colorectal cancer.


Asunto(s)
Calgranulina A/fisiología , Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Hormonas Tiroideas/fisiología , Proteínas 14-3-3 , Calgranulina A/genética , Calgranulina B/genética , Calgranulina B/fisiología , Proteínas Portadoras/genética , Línea Celular Tumoral , Cromatografía Liquida , Neoplasias Colorrectales/genética , Células HEK293 , Humanos , Inmunoprecipitación , Proteínas de la Membrana/genética , Nucleofosmina , Mapeo de Interacción de Proteínas , Espectrometría de Masas en Tándem , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
17.
Biochem Biophys Res Commun ; 484(1): 184-188, 2017 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-28088518

RESUMEN

Airway hyperresponsiveness (AHR) is a major clinical problem in allergic asthma mainly caused by the hypercontractility of airway smooth muscles (ASM). S100A8 is an important member of the S100 calcium-binding protein family with a potential to regulate cell contractility. Here, we analyze the potential of S100A8 to regulate allergen-induced AHR and ASM contraction. Treatment with recombinant S100A8 (rS100A8) diminished airway hyperresponsiveness in OVA-sensitized rats. ASM contraction assays showed that rS100A8 reduced hypercontractility in both isolated tracheal rings and primary ASM cells treated by acetylcholine. rS100A8 markedly rescued the phosphorylation level of myosin light chain induced by acetylcholine in ASM cells. These results show that rS100A8 plays a protective role in regulating AHR in asthma by inhibiting ASM contraction. These results support S100A8 as a novel therapeutic target to control ASM contraction in asthma.


Asunto(s)
Calgranulina A/fisiología , Músculo Liso/fisiología , Hipersensibilidad Respiratoria/prevención & control , Acetilcolina/administración & dosificación , Animales , Células Cultivadas , Contracción Muscular/fisiología , Cadenas Ligeras de Miosina/metabolismo , Ovalbúmina/administración & dosificación , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología
18.
J Gerontol A Biol Sci Med Sci ; 72(3): 319-328, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27131040

RESUMEN

Inflammation plays an important role in Alzheimer's disease (AD) and other neurodegenerative disorders. Although chronic inflammation in later stages of AD is well described, little is known about the inflammatory processes in preclinical or early stages of the disease prior to plaque deposition. In this study, we report that the inflammatory mediator S100A8 is increased with aging in the mouse brain. It is observed as extracellular aggregates, which do not correspond to corpora amylacea. S100A8 aggregation is enhanced in the hippocampi of two different mouse models for amyloid-ß (Aß) overproduction (Tg2576 and TgAPParctic mice). S100A8 aggregates are seen prior the formation of Aß plaques and do not colocalize. In vitro treatment of glial cells from primary cultures with Aß42 resulted in an increased production of S100A8. In parallel, treatment of a neuronal cell line with recombinant S100A8 protein resulted in enhanced Aß42 and decreased Aß40 production. Our results suggest that important inflammatory processes are occurring prior to Aß deposition and the existence of a positive feedback between S100A8 and Aß productions. The possible relevance of aging- or AD-dependent formation of S100A8 aggregates in the hippocampus thus affecting learning and memory processes is discussed.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/fisiología , Calgranulina A/fisiología , Retroalimentación Fisiológica , Hipocampo/patología , Inflamación/etiología , Agregación Patológica de Proteínas , Factores de Edad , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/biosíntesis , Animales , Calgranulina A/metabolismo , Hipocampo/metabolismo , Ratones , Ratones Transgénicos
19.
Curr Diab Rep ; 16(10): 100, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27612847

RESUMEN

The receptor for advanced glycation end products (RAGE) is a novel protein increasingly studied in the pathogenesis of type 1 diabetes (T1D). RAGE is expressed by several immune cell types, including T cells, antigen-presenting cells, endothelial cells, and the endocrine cells of the pancreatic islets. RAGE binds various ligands including advanced glycation end products (AGEs), high-mobility group box protein 1 (HMGB1), S100 proteins, ß-amyloid, ß-sheet fibrils, and lipopolysaccharide. AGEs are a particularly interesting ligand because their exogenous introduction into the body can be accelerated by the consumption of AGE-rich processed foods. This review will detail RAGE isoforms and its ligands and discuss how RAGE binding on the aforementioned cells could be linked to T1D pathogenesis.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Inmunidad Adaptativa , Animales , Calgranulina A/fisiología , Productos Finales de Glicación Avanzada/metabolismo , Proteína HMGB1/fisiología , Humanos , Inmunidad Innata , Islotes Pancreáticos/fisiología
20.
Biochem Biophys Res Commun ; 472(1): 243-9, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26920052

RESUMEN

S100A8 is an important member of the S100 protein family, which is involved in intracellular and extracellular regulatory activities. We previously reported that the S100A8 protein was differentially expressed in the asthmatic respiratory tracts. To understand the potential role of S100A8 in asthma, we investigated the effect of recombinant S100A8 protein on the platelet-derived growth factor (PDGF)-induced migration of airway smooth muscle cells (ASMCs) and the underlying molecular mechanism by using multiple methods, such as impedance-based xCELLigence migration assay, transwell migration assays and wound-healing assays. We found that exogenous S100A8 protein significantly inhibited PDGF-induced ASMC migration. Furthermore, the migration inhibition effect of S100A8 was blocked by neutralizing antibody against the receptor for advanced glycation end-products (RAGE), a potential receptor for the S100A8 protein. These findings provide direct evidence that exogenous S100A8 protein inhibits the PDGF-induced migration of ASMCs through the membrane receptor RAGE. Our study highlights a novel role of S100A8 as a potential means of counteracting airway remodeling in chronic airway diseases.


Asunto(s)
Calgranulina A/fisiología , Movimiento Celular/fisiología , Miocitos del Músculo Liso/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Tráquea/patología , Tráquea/fisiología , Animales , Anticuerpos Neutralizantes , Asma/patología , Asma/fisiopatología , Calgranulina A/administración & dosificación , Calgranulina A/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ratas , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA