Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(7)2020 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-32235291

RESUMEN

Regulatory T cells (Tregs) are important mediators of immunological self-tolerance and homeostasis. Being cluster of differentiation 4+Forkhead box protein3+ (CD4+FOXP3+), these cells are a subset of CD4+ T lymphocytes and can originate from the thymus (tTregs) or from the periphery (pTregs). The malfunction of CD4+ Tregs is associated with autoimmune responses such as rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), inflammatory bowel diseases (IBD), psoriasis, systemic lupus erythematosus (SLE), and transplant rejection. Recent evidence supports an opposed role in sepsis. Therefore, maintaining functional Tregs is considered as a therapy regimen to prevent autoimmunity and allograft rejection, whereas blocking Treg differentiation might be favorable in sepsis patients. It has been shown that Tregs can be generated from conventional naïve T cells, called iTregs, due to their induced differentiation. Moreover, Tregs can be effectively expanded in vitro based on blood-derived tTregs. Taking into consideration that the suppressive role of Tregs has been mainly attributed to the expression and function of the transcription factor Foxp3, modulating its expression and binding to the promoter regions of target genes by altering the chromatin histone acetylation state may turn out beneficial. Hence, we discuss the role of histone deacetylation inhibitors as epigenetic modulators of Tregs in this review in detail.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Inmunomodulación/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/fisiología , Animales , Autoinmunidad , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Susceptibilidad a Enfermedades , Epigénesis Genética , Histona Desacetilasas/metabolismo , Humanos , Tolerancia Inmunológica , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología
2.
Int J Mol Sci ; 21(20)2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33065981

RESUMEN

During the course of sepsis in critically ill patients, kidney dysfunction and damage are among the first events of a complex scenario toward multi-organ failure and patient death. Acute kidney injury triggers the release of lipocalin-2 (Lcn-2), which is involved in both renal injury and recovery. Taking into account that Lcn-2 binds and transports iron with high affinity, we aimed at clarifying if Lcn-2 fulfills different biological functions according to its iron-loading status and its cellular source during sepsis-induced kidney failure. We assessed Lcn-2 levels both in serum and in the supernatant of short-term cultured renal macrophages (MΦ) as well as renal tubular epithelial cells (TEC) isolated from either Sham-operated or cecal ligation and puncture (CLP)-treated septic mice. Total kidney iron content was analyzed by Perls' staining, while Lcn-2-bound iron in the supernatants of short-term cultured cells was determined by atomic absorption spectroscopy. Lcn-2 protein in serum was rapidly up-regulated at 6 h after sepsis induction and subsequently increased up to 48 h. Lcn-2-levels in the supernatant of TEC peaked at 24 h and were low at 48 h with no change in its iron-loading. In contrast, in renal MΦ Lcn-2 was low at 24 h, but increased at 48 h, where it mainly appeared in its iron-bound form. Whereas TEC-secreted, iron-free Lcn-2 was associated with renal injury, increased MΦ-released iron-bound Lcn-2 was linked to renal recovery. Therefore, we hypothesized that both the cellular source of Lcn-2 as well as its iron-load crucially adds to its biological function during sepsis-induced renal injury.


Asunto(s)
Hierro/metabolismo , Lipocalina 2/metabolismo , Insuficiencia Renal/metabolismo , Sepsis/complicaciones , Animales , Biomarcadores/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Lipocalina 2/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Insuficiencia Renal/etiología , Insuficiencia Renal/patología
3.
Bioorg Med Chem ; 27(21): 115082, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31548084

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) modulators have found wide application for the treatment of cancers, metabolic disorders and inflammatory diseases. Contrary to PPARγ agonists, PPARγ antagonists have been much less studied and although they have shown immunomodulatory effects, there is still no therapeutically useful PPARγ antagonist on the market. In contrast to non-competitive, irreversible inhibition caused by 2-chloro-5-nitrobenzanilide (GW9662), the recently described (E)-2-(5-((4-methoxy-2-(trifluoromethyl)quinolin-6-yl)methoxy)-2-((4-(trifluoromethyl)benzyl)oxy)-benzylidene)-hexanoic acid (MTTB, T-10017) is a promising prototype for a new class of PPARγ antagonists. It exhibits competitive antagonism against rosiglitazone mediated activation of PPARγ ligand binding domain (PPARγLBD) in a transactivation assay in HEK293T cells with an IC50 of 4.3 µM against 1 µM rosiglitazone. The aim of this study was to investigate the structure-activity relationships (SAR) of the MTTB scaffold focusing on improving its physicochemical properties. Through this optimization, 34 new derivatives were prepared and characterized. Two new potent compounds (T-10075 and T-10106) with much improved drug-like properties and promising pharmacokinetic profile were identified.


Asunto(s)
Cinamatos/farmacología , PPAR gamma/antagonistas & inhibidores , Quinolinas/farmacología , Animales , Cinamatos/síntesis química , Cinamatos/farmacocinética , Células HEK293 , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/farmacocinética , Ratas , Rosiglitazona/farmacología , Relación Estructura-Actividad
4.
Int J Mol Sci ; 20(2)2019 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-30654448

RESUMEN

Sepsis is characterized by dysregulated gene expression, provoking a hyper-inflammatory response occurring in parallel to a hypo-inflammatory reaction. This is often associated with multi-organ failure, leading to the patient's death. Therefore, reprogramming of these pro- and anti-inflammatory, as well as immune-response genes which are involved in acute systemic inflammation, is a therapy approach to prevent organ failure and to improve sepsis outcomes. Considering epigenetic, i.e., reversible, modifications of chromatin, not altering the DNA sequence as one tool to adapt the expression profile, inhibition of factors mediating these changes is important. Acetylation of histones by histone acetyltransferases (HATs) and initiating an open-chromatin structure leading to its active transcription is counteracted by histone deacetylases (HDACs). Histone deacetylation triggers a compact nucleosome structure preventing active transcription. Hence, inhibiting the activity of HDACs by specific inhibitors can be used to restore the expression profile of the cells. It can be assumed that HDAC inhibitors will reduce the expression of pro-, as well as anti-inflammatory mediators, which blocks sepsis progression. However, decreased cytokine expression might also be unfavorable, because it can be associated with decreased bacterial clearance.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Epigénesis Genética , Humanos , Modelos Biológicos , Sepsis/genética
5.
Lab Invest ; 98(11): 1408-1422, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30087458

RESUMEN

Macrophage-epithelial cross-talk regulates cell cycle progression and represents an important factor in rescuing epithelial cells from cell cycle arrest in order to maintain a healthy epithelial phenotype. However, the underlying mechanisms are still not well defined. We provide evidence that macrophage-secreted lipocalin-2 (Lcn-2) plays a key role during this process. In a co-culture setup using cell cycle arrested NRK52e renal epithelial cells and primary bone marrow-derived macrophages, Lcn-2 restores proliferation through inhibition of peroxisome proliferator-activated receptor (PPAR)-γ. Lcn-2 overexpression in macrophages overcomes epithelial cell cycle arrest and enhances epithelial markers via megalin and the downstream activation of PI3K/Akt signalling pathway, whereas a knockdown of Lcn-2 in macrophages prevented this effect. Our results show that macrophage-secreting Lcn-2 is crucial in rescuing epithelial cells from cell cycle arrest and in promoting epithelial proliferation.


Asunto(s)
Proliferación Celular , Células Epiteliales/fisiología , Lipocalina 2/fisiología , PPAR gamma/metabolismo , Animales , Ciclo Celular , Línea Celular , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley
7.
Int J Mol Sci ; 18(12)2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29261130

RESUMEN

Tight regulation of inflammation is very important to guarantee a balanced immune response without developing chronic inflammation. One of the major mediators of the resolution of inflammation is the transcription factor: the nuclear factor erythroid 2-like 2 (Nrf2). Stabilized following oxidative stress, Nrf2 induces the expression of antioxidants as well as cytoprotective genes, which provoke an anti-inflammatory expression profile, and is crucial for the initiation of healing. In view of this fundamental modulatory role, it is clear that both hyper- or hypoactivation of Nrf2 contribute to the onset of chronic diseases. Understanding the tight regulation of Nrf2 expression/activation and its interaction with signaling pathways, known to affect inflammatory processes, will facilitate development of therapeutic approaches to prevent Nrf2 dysregulation and ameliorate chronic inflammatory diseases. We discuss in this review the principle mechanisms of Nrf2 regulation with a focus on inflammation and autophagy, extending the role of dysregulated Nrf2 to chronic diseases and tumor development.


Asunto(s)
Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Animales , Antioxidantes/farmacología , Autofagia , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/genética
8.
J Biol Chem ; 290(47): 28446-28455, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26459563

RESUMEN

The activation of the transcription factor NF-E2-related factor 2 (Nrf2) maintains cellular homeostasis in response to oxidative stress by the regulation of multiple cytoprotective genes. Without stressors, the activity of Nrf2 is inhibited by its interaction with the Keap1 (kelch-like ECH-associated protein 1). Here, we describe (3S)-1-[4-[(2,3,5,6-tetramethylphenyl) sulfonylamino]-1-naphthyl]pyrrolidine-3-carboxylic acid (RA839), a small molecule that binds noncovalently to the Nrf2-interacting kelch domain of Keap1 with a Kd of ∼6 µM, as demonstrated by x-ray co-crystallization and isothermal titration calorimetry. Whole genome DNA arrays showed that at 10 µM RA839 significantly regulated 105 probe sets in bone marrow-derived macrophages. Canonical pathway mapping of these probe sets revealed an activation of pathways linked with Nrf2 signaling. These pathways were also activated after the activation of Nrf2 by the silencing of Keap1 expression. RA839 regulated only two genes in Nrf2 knock-out macrophages. Similar to the activation of Nrf2 by either silencing of Keap1 expression or by the reactive compound 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me), RA839 prevented the induction of both inducible nitric-oxide synthase expression and nitric oxide release in response to lipopolysaccharides in macrophages. In mice, RA839 acutely induced Nrf2 target gene expression in liver. RA839 is a selective inhibitor of the Keap1/Nrf2 interaction and a useful tool compound to study the biology of Nrf2.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Pirrolidinas/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Proteína 1 Asociada A ECH Tipo Kelch , Masculino , Ratones , Unión Proteica , Pirrolidinas/metabolismo , Sulfonamidas/metabolismo
9.
Am J Respir Cell Mol Biol ; 53(5): 656-63, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25860173

RESUMEN

Nerve injury-induced protein (Ninjurin [Ninj]) 1 is an adhesion molecule originally identified in Schwann cells after nerve injury, whereas it is also expressed in leukocytes, epithelium, endothelium, and various organs, and is induced under inflammatory conditions. Its contribution to inflammation was so far restricted to the nervous system and exclusively attributed to its role during leukocyte migration. We hypothesized a proinflammatory role for Ninj1 also outside the nervous system. To elucidate its impact during inflammation, we analyzed expression levels and its contribution to inflammation in septic mice and studied its effect on inflammatory signaling in vitro. The effect on inflammation was analyzed by genetic (only in vitro) and pharmacologic repression in septic mice (cecal ligation and puncture) and cell culture, respectively. Repression of Ninj1 by an inhibitory peptide or small interfering RNA attenuated LPS-triggered inflammation in macrophages and endothelial cells by modulating p38 phosphorylation and activator protein-1 activation. Inhibition of Ninj1 in septic mice reduced systemic and pulmonary inflammation as well as organ damage, and ameliorated survival after 24 hours. Ninj1 is elevated under inflammatory conditions and contributes to inflammation not only by mediating leukocyte migration, but also by modulating Toll-like receptor 4-dependent expression of inflammatory mediators. We assume that, owing to both mechanisms, inhibition reduces systemic inflammation and organ damage in septic mice. Our data contribute to a better understanding of the complex inflammatory mechanisms and add a novel therapeutic target for inflammatory conditions such as sepsis.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/inmunología , Factores de Crecimiento Nervioso/inmunología , Sepsis/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Receptor Toll-Like 4/inmunología , Animales , Moléculas de Adhesión Celular Neuronal/genética , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/patología , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/genética , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Cultivo Primario de Células , Sepsis/genética , Sepsis/patología , Transducción de Señal , Síndrome de Respuesta Inflamatoria Sistémica/genética , Síndrome de Respuesta Inflamatoria Sistémica/patología , Ácidos Teicoicos/farmacología , Receptor Toll-Like 4/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
11.
Blood Adv ; 8(11): 2870-2879, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38588481

RESUMEN

ABSTRACT: Hemojuvelin (HJV) is a glycosylphosphatidylinositol-anchored protein of the repulsive guidance molecule family acting as a bone morphogenetic protein (BMP) coreceptor to induce the hepatic iron regulatory protein hepcidin. Hepcidin causes ubiquitination and degradation of the sole known iron exporter ferroportin, thereby limiting iron availability. The detailed signaling mechanism of HJV in vivo has yet to be investigated. In the current manuscript, we used an established model of adeno-associated virus (AAV)-mediated liver-specific overexpression of HJV in murine models of hepatocyte-specific deficiency of the BMP type I receptors Alk2 or Alk3. In control mice, HJV overexpression increased hepatic Hamp messenger RNA (mRNA) levels, soluble HJV (sHJV), splenic iron content (SIC), as well as phosphorylated small mothers against decapentaplegic protein (pSMAD1/5/8) levels. In contrast, in Alk2fl/fl;Alb-Cre and Alk3fl/fl;Alb-Cre mice, which present with moderate and severe iron overload, respectively, the administration of AAV-HJV induced HJV and sHJV. However, it did not rescue the iron overload phenotypes of those mice. Serum iron levels were induced in Alk2fl/fl;Alb-Cre mice after HJV overexpression. In phosphate-buffered saline-injected Alk3fl/fl;Alb-Cre mice, serum iron levels and the expression of duodenal ferroportin remained high, whereas Hamp mRNA levels were decreased to 1% to 5% of the levels detected in controls. This was reduced even further by AAV-HJV overexpression. SIC remained low in mice with hepatocyte-specific Alk2 or Alk3 deficiency, reflecting disturbed iron homeostasis with high serum iron levels and transferrin saturation and an inability to induce hepcidin by HJV overexpression. The data indicate that ALK2 and ALK3 are both required in vivo for the HJV-mediated induction of hepcidin.


Asunto(s)
Proteínas Ligadas a GPI , Proteína de la Hemocromatosis , Hepcidinas , Animales , Ratones , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/genética , Hepcidinas/metabolismo , Hepcidinas/genética , Proteína de la Hemocromatosis/metabolismo , Proteína de la Hemocromatosis/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Hígado/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II
12.
Exp Cell Res ; 318(19): 2490-7, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22884583

RESUMEN

Heterodimerization and cross-talk between nuclear hormone receptors often occurs. For example, estrogen receptor alpha (ERα) physically binds to peroxisome proliferator-activated receptor gamma (PPARγ) and inhibits its transcriptional activity. The interaction between PPARγ and the vitamin D receptor (VDR) however, is unknown. Here, we elucidate the molecular mechanisms linking PPARγ and VDR signaling, and for the first time we show that PPARγ physically associates with VDR in human breast cancer cells. We found that overexpression of PPARγ decreased 1α,25-dihydroxyvitamin D(3) (1,25D(3)) mediated transcriptional activity of the vitamin D target gene, CYP24A1, by 49% and the activity of VDRE-luc, a vitamin D responsive reporter, by 75% in T47D human breast cancer cells. Deletion mutation experiments illustrated that helices 1 and 4 of PPARγ's hinge and ligand binding domains, respectively, governed this suppressive function. Additionally, abrogation of PPARγ's AF2 domain attenuated its repressive action on 1,25D(3) transactivation, indicating that this domain is integral in inhibiting VDR signaling. PPARγ was also found to compete with VDR for their binding partner retinoid X receptor alpha (RXRα). Overexpression of RXRα blocked PPARγ's suppressive effect on 1,25D(3) action, enhancing VDR signaling. In conclusion, these observations uncover molecular mechanisms connecting the PPARγ and VDR pathways.


Asunto(s)
Neoplasias de la Mama/genética , Colestanotriol 26-Monooxigenasa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Receptor Cross-Talk/fisiología , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Activación Transcripcional , Neoplasias de la Mama/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Colestanotriol 26-Monooxigenasa/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Células MCF-7 , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptor alfa X Retinoide/genética , Receptor alfa X Retinoide/metabolismo , Eliminación de Secuencia , Transducción de Señal , Esteroide Hidroxilasas/genética , Esteroide Hidroxilasas/metabolismo , Células Tumorales Cultivadas , Vitamina D3 24-Hidroxilasa
13.
Shock ; 60(6): 727-738, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37878473

RESUMEN

ABSTRACT: Background: Severe progression of COVID-19 to critical illness, with pulmonary failure, multiple organ failure, and death, is driven by systemic inflammatory responses with overproduction of inflammatory cytokines. In the past years, the potential role of bradykinin, leading to inappropriate immune responses in the pathogenesis of COVID-19, has been raised in a so-called bradykinin storm. However, clinical investigations of bradykinin, its metabolite des-Arg 9 -bradykinin, or substance P, are rare or completely lacking during intensive care of COVID-19 patients. A prospective prolonged cohort study was conducted, including 44 COVID-19 patients (09/2020-02/2021, prevalent wildtype SARS-CoV-2) from the intensive care unit. Plasma levels of bradykinin, des-Arg 9 -bradykinin, and substance P were measured daily by ELISA in survivors (n = 21) and nonsurvivors (n = 23) of COVID-19 from admission until discharge or death. Results: We found significantly higher plasma levels of des-Arg 9 -bradykinin in survivors and nonsurvivors of COVID-19 compared with healthy controls. In addition, plasma des-Arg 9 -bradykinin levels were higher ( P < 0.001, effect size = 0.79) in nonsurvivors compared with survivors of COVID-19 and correlated significantly with disease worsening, and clinical parameters of inflammation, like leukocyte count, IL-6 or lactate dehydrogenase, and outcome. Consequently, compared with healthy controls, bradykinin and substance P plasma levels were significantly reduced in survivors and nonsurvivors of COVID-19. Furthermore, plasma substance P levels were significantly reduced ( P < 0.001, effect size = 0.7) in nonsurvivors compared with survivors of COVID-19, whereas plasma bradykinin levels did not significantly differ between survivors and nonsurvivors of COVID-19. Conclusion: Our data demonstrates that des-Arg 9 -bradykinin is significantly elevated in COVID-19 intensive care unit patients and is associated with disease severity, clinical inflammatory parameters, and survival. These results indicate that des-Arg 9 -bradykinin, not bradykinin, is one of the pivotal peptides of concern for the lethal COVID-19 aggravation and outcome. Further investigations are necessary to evaluate whether des-Arg 9 -bradykinin exhibits potent blood biomarker properties in COVID-19 and offer new treatment approaches.


Asunto(s)
Bradiquinina , COVID-19 , Humanos , Receptores de Bradiquinina/metabolismo , Estudios de Cohortes , Estudios Prospectivos , Sustancia P , SARS-CoV-2/metabolismo
14.
Front Immunol ; 14: 1240394, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38322012

RESUMEN

Background: Interactions between tumor cells and cells in the microenvironment contribute to tumor development and metastasis. The spatial arrangement of individual cells in relation to each other influences the likelihood of whether and how these cells interact with each other. Methods: This study investigated the effect of spatial distribution on the function of leukocyte subsets in the microenvironment of human head and neck squamous cell carcinoma (HNSCC) using multiplex immunohistochemistry (IHC). Leukocyte subsets were further classified based on analysis of two previously published HNSCC single-cell RNA datasets and flow cytometry (FC). Results: IHC revealed distinct distribution patterns of leukocytes differentiated by CD68 and CD163. While CD68hiCD163lo and CD68hiCD163hi cells accumulated near tumor sites, CD68loCD163hi cells were more evenly distributed in the tumor stroma. PD-L1hi and PD-1hi cells accumulated predominantly around tumor sites. High cell density of PD-L1hi CD68hiCD163hi cells or PD-1hi T cells near the tumor site correlated with improved survival. FC and single cell RNA revealed high variability within the CD68/CD163 subsets. CD68hiCD163lo and CD68hiCD163hi cells were predominantly macrophages (MΦ), whereas CD68loCD163hi cells appeared to be predominantly dendritic cells (DCs). Differentiation based on CD64, CD80, CD163, and CD206 revealed that TAM in HNSCC occupy a broad spectrum within the classical M1/M2 polarization. Notably, the MΦ subsets expressed predominantly CD206 and little CD80. The opposite was observed in the DC subsets. Conclusion: The distribution patterns and their distinct interactions via the PD-L1/PD-1 pathway suggest divergent roles of CD68/CD163 subsets in the HNSCC microenvironment. PD-L1/PD-1 interactions appear to occur primarily between specific cell types close to the tumor site. Whether PD-L1/PD-1 interactions have a positive or negative impact on patient survival appears to depend on both the spatial localization and the entity of the interacting cells. Co-expression of other markers, particularly CD80 and CD206, supports the hypothesis that CD68/CD163 IHC subsets have distinct functions. These results highlight the association between spatial leukocyte distribution patterns and the clinical presentation of HNSCC.


Asunto(s)
Antígeno B7-H1 , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1 , Linfocitos T/metabolismo , ARN , Microambiente Tumoral
15.
J Cell Sci ; 123(Pt 2): 192-201, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20026644

RESUMEN

PPARgamma exerts significant anti-inflammatory signaling properties in monocytes and macrophages, which are affected by its intracellular localization. Based on our previous report, which showed that cytosolic localization of PPARgamma attenuates PKCalpha signaling in macrophages, we elucidated the molecular mechanisms provoking cytosolic PPARgamma localization. Using the DsRed-tagged PPARgamma deletion constructs PPARgamma1 Delta1-31 and PPARgamma1 Delta407-475, we observed an exclusive nuclear PPARgamma1 Delta1-31 localization in transfected HEK293 cells, whereas PPARgamma1 Delta407-475 did not alter its cytosolic or nuclear localization. The casein kinase II (CK-II) inhibitor 5,6-dichloro-1-beta-D-ribofuranosyl benzimidazole (DRB) prevented cytosolic PPARgamma localization. Mutation of two possible CK-II phosphorylation sites at serine 16 and serine 21 of PPARgamma into alanine (PPARgamma S16A/S21A) inhibited cytosolic PPARgamma localization. Moreover, a PPARgamma S16E/S21E mutant that mimicks constitutive phosphorylation of residues 16 and 21, predominantly resides in the cytosol. The CRM1 inhibitor leptomycin B abolished cytosolic PPARgamma localization, suggesting that this is a CRM1-dependent export process. CRM1-mediated PPARgamma export requires Ran and phosphorylated RanBP3. Finally, co-immunoprecipitation studies demonstrated that DRB blocks PPARgamma binding to CRM1, whereas PD98059 inhibits RanBP3 binding to CRM1 and concomitant shuttling from nucleus to cytosol, but does not alter PPARgamma binding to CRM1. We conclude that CK-II-dependent PPARgamma phosphorylation at Ser16 and Ser21 is necessary for CRM1/Ran/RanBP3-mediated nucleocytoplasmic translocation of PPARgamma.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Carioferinas/metabolismo , PPAR gamma/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Núcleo Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Ácidos Grasos Insaturados/farmacología , Técnicas de Silenciamiento del Gen , Ratones , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Compuestos Orgánicos/farmacología , PPAR gamma/química , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteína de Unión al GTP ran/metabolismo , Proteína Exportina 1
16.
Blood ; 115(17): 3531-40, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20197547

RESUMEN

Execution of physiologic cell death known as apoptosis is tightly regulated and transfers immunologically relevant information. This ensures efficient clearance of dying cells and shapes the phenotype of their "captors" toward anti-inflammatory. Here, we identify a mechanism of sphingosine-1-phosphate production by apoptotic cells. During cell death, sphingosine kinase 2 (SphK2) is cleaved at its N-terminus in a caspase-1-dependent manner. Thereupon, a truncated but enzymatically active fragment of SphK2 is released from cells. This step is coupled to phosphatidylserine exposure, which is a hallmark of apoptosis and a crucial signal for phagocyte/apoptotic cell interaction. Our data link signaling events during apoptosis to the extracellular production of a lipid mediator that affects immune cell attraction and activation.


Asunto(s)
Apoptosis/inmunología , Caspasa 1/inmunología , Lisofosfolípidos/inmunología , Fosfotransferasas (Aceptor de Grupo Alcohol)/inmunología , Esfingosina/análogos & derivados , Animales , Caspasa 1/genética , Humanos , Células Jurkat , Lisofosfolípidos/metabolismo , Ratones , Células 3T3 NIH , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/inmunología , Esfingosina/metabolismo
17.
J Cell Biol ; 176(5): 681-94, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17325208

RESUMEN

Recently, we provided evidence that PKCalpha depletion in monocytes/macrophages contributes to cellular desensitization during sepsis. We demonstrate that peroxisome proliferator-activated receptor gamma (PPARgamma) agonists dose dependently block PKCalpha depletion in response to the diacylglycerol homologue PMA in RAW 264.7 and human monocyte-derived macrophages. In these cells, we observed PPARgamma-dependent inhibition of nuclear factor-kappaB (NF-kappaB) activation and TNF-alpha expression in response to PMA. Elucidating the underlying mechanism, we found PPARgamma1 expression not only in the nucleus but also in the cytoplasm. Activation of PPARgamma1 wild type, but not an agonist-binding mutant of PPARgamma1, attenuated PMA-mediated PKCalpha cytosol to membrane translocation. Coimmunoprecipitation assays pointed to a protein-protein interaction of PKCalpha and PPARgamma1, which was further substantiated using a mammalian two-hybrid system. Applying PPARgamma1 mutation and deletion constructs, we identified the hinge helix 1 domain of PPARgamma1 that is responsible for PKCalpha binding. Therefore, we conclude that PPARgamma1-dependent inhibition of PKCalpha translocation implies a new model of macrophage desensitization.


Asunto(s)
Membrana Celular/enzimología , Macrófagos/enzimología , PPAR gamma/fisiología , Proteína Quinasa C-alfa/metabolismo , Animales , Sitios de Unión , Biomarcadores/análisis , Células COS , Chlorocebus aethiops , Citosol/enzimología , Humanos , Modelos Biológicos , Mutación , PPAR gamma/química , PPAR gamma/genética , Proteína Quinasa C-alfa/análisis , Proteína Quinasa C-alfa/química , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Transducción de Señal , Técnicas del Sistema de Dos Híbridos
18.
Cell Mol Life Sci ; 68(10): 1815-27, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20949368

RESUMEN

Apoptotic cell (AC)-derived factors alter the physiology of macrophages (MΦs) towards a regulatory phenotype, characterized by reduced nitric oxide (NO) production. Impaired NO formation in response to AC-conditioned medium (CM) was facilitated by arginase II (ARG II) expression, which competes with inducible NO synthase for L-arginine. Here we explored signaling pathways allowing CM to upregulate ARG II in RAW264.7 MΦs. Sphingosine-1-phosphate (S1P) was required and acted synergistically with a so far unidentified factor to elicit high ARG II expression. S1P activated S1P(2), since S1P(2) knockdown prevented ARG II upregulation. Furthermore, ERK5 knockdown attenuated CM-mediated ARG II protein induction. CREB was implicated as shown by EMSA analysis and decoy-oligonucleotides scavenging CREB in RAW264.7 MΦs, which blocked ARG II expression. We conclude that AC-derived S1P binds to S1P(2) and acts synergistically with other factors to activate ERK5 and concomitantly CREB. This signaling cascade shapes an anti-inflammatory MΦ phenotype by ARG II induction.


Asunto(s)
Arginasa/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Macrófagos/enzimología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Animales , Apoptosis , Línea Celular , Medios de Cultivo Condicionados/farmacología , Ensayo de Cambio de Movilidad Electroforética , Humanos , Lisofosfolípidos/farmacología , Ratones , Proteína Quinasa 7 Activada por Mitógenos/genética , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología
19.
Am J Respir Crit Care Med ; 184(1): 64-74, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21471100

RESUMEN

RATIONALE: Despite intensive research, sepsis displays the most prevalent cause of death on intensive care units. The hallmark of sepsis is an overshooting T-cell death that reduces host defense mechanisms and that is associated with poor patient survival. Previous in vitro studies revealed that the expression of the transcription factor peroxisome proliferator-activated receptor (PPAR) γ was increased in isolated T cells of patients with sepsis. OBJECTIVES: We determined the importance of targeting PPARγ for sepsis treatment and underlying molecular mechanisms for T-cell apoptosis in vivo. METHODS: To mimic human systemic inflammation and septic conditions, we used a nonlethal endotoxemia and a lethal cecum ligation and puncture polymicrobial sepsis model. MEASUREMENTS AND MAIN RESULTS: PPARγ inhibition in T cells with either the PPARγ antagonist GW9662 or a newly generated T cell-specific PPARγ knockout (Tc-PPARγ(-/-)) mice provided a survival advantage during polymicrobial sepsis in mice, which correlated with abrogated T-cell depletion in both in vivo models. Pathway analysis revealed increased antiapoptotic IL-2 and Bcl-2 expression, and activated prosurvival PI3K/Akt signaling under PPARγ-deficient conditions. In line, neutralizing IL-2 in Tc-PPARγ(-/-) mice resulted in T-cell apoptosis and increased mortality. CONCLUSIONS: Our results provide evidence for a pivotal involvement of PPARγ in T-cell depletion by activating two important apoptosis pathways, and subsequently provoking the breakdown of defense mechanisms during systemic inflammation and sepsis.


Asunto(s)
Apoptosis , PPAR gamma/fisiología , Sepsis/mortalidad , Linfocitos T/fisiología , Anilidas/farmacología , Animales , Interleucina-2/metabolismo , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , PPAR gamma/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Peritonitis/inmunología , Peritonitis/microbiología , Peritonitis/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sepsis/inmunología , Sepsis/microbiología , Sepsis/fisiopatología , Transducción de Señal , Tasa de Supervivencia
20.
Biomedicines ; 10(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35052778

RESUMEN

Acute respiratory distress syndrome (ARDS) is a major cause of patient mortality in intensive care units (ICUs) worldwide. Considering that no causative treatment but only symptomatic care is available, it is obvious that there is a high unmet medical need for a new therapeutic concept. One reason for a missing etiologic therapy strategy is the multifactorial origin of ARDS, which leads to a large heterogeneity of patients. This review summarizes the various kinds of ARDS onset with a special focus on the role of reactive oxygen species (ROS), which are generally linked to ARDS development and progression. Taking a closer look at the data which already have been established in mouse models, this review finally proposes the translation of these results on successful antioxidant use in a personalized approach to the ICU patient as a potential adjuvant to standard ARDS treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA