Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Bull Exp Biol Med ; 174(4): 544-548, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36894814

RESUMEN

We studied the influence of activated innate and adaptive immune cells on the production of growth factors by human adipose tissue multipotent mesenchymal stromal cells (MSC). MSC showed immunosuppressive properties in vitro: decreased activation and proliferation of stimulated immune cells. T-cell interaction with MSC resulted with increased secretion of EGF, PDGF-AB/BB, FGF-2, and VEGF growth factors. Co-culturing with natural killer cells also stimulated TGFα production. The intensity of the effect varied depending on the type of immune cells. Natural killer caused a more significant increase in PDGF-AB/BB and FGF-2 secretion, while VEGF secretion increased stronger after co-culturing with T cells. The obtained data indicate the possibility of increasing reparative potential of MSC under the influence of inflammatory microenvironment.


Asunto(s)
Microambiente Celular , Inflamación , Células Madre Mesenquimatosas , Humanos , Becaplermina , Proliferación Celular , Microambiente Celular/inmunología , Técnicas de Cocultivo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor A de Crecimiento Endotelial Vascular , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Comunicación Paracrina/inmunología , Inflamación/inmunología , Inflamación/metabolismo
2.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34847076

RESUMEN

Despite decades of research, there is no specific therapy for acute pancreatitis (AP). In the current study, we have evaluated the efficacy of pirfenidone, an antiinflammatory and antifibrotic agent that is approved by the FDA for treatment of idiopathic pulmonary fibrosis (IPF), in ameliorating local and systemic injury in AP. Our results suggest that treatment with pirfenidone in therapeutic settings (e.g., after initiation of injury), even when administered at the peak of injury, reduces severity of local and systemic injury and inflammation in multiple models of AP. In vitro evaluation suggests that pirfenidone decreases cytokine release from acini and macrophages and disrupts acinar-macrophage crosstalk. Therapeutic pirfenidone treatment increases IL-10 secretion from macrophages preceding changes in histology and modulates the immune phenotype of inflammatory cells with decreased levels of inflammatory cytokines. Antibody-mediated IL-10 depletion, use of IL-10-KO mice, and macrophage depletion experiments confirmed the role of IL-10 and macrophages in its mechanism of action, as pirfenidone was unable to reduce severity of AP in these scenarios. Since pirfenidone is FDA approved for IPF, a trial evaluating the efficacy of pirfenidone in patients with moderate to severe AP can be initiated expeditiously.


Asunto(s)
Células Acinares/metabolismo , Fibrosis , Interleucina-10/inmunología , Macrófagos/metabolismo , Páncreas , Pancreatitis , Piridonas/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Células Cultivadas , Citocinas/clasificación , Citocinas/inmunología , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/prevención & control , Ratones , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/lesiones , Páncreas/patología , Pancreatitis/tratamiento farmacológico , Pancreatitis/inmunología , Comunicación Paracrina/inmunología , Transducción de Señal/inmunología
3.
Gastroenterology ; 162(2): 590-603.e14, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34627860

RESUMEN

BACKGROUND AND AIMS: Patients with pancreatic ductal adenocarcinoma (PDA) have not yet benefitted from the revolution in cancer immunotherapy due in large part to a dominantly immunosuppressive tumor microenvironment. MEK inhibition combined with autophagy inhibition leads to transient tumor responses in some patients with PDA. We examined the functional effects of combined MEK and autophagy inhibition on the PDA immune microenvironment and the synergy of combined inhibition of MEK and autophagy with CD40 agonism (aCD40) against PDA using immunocompetent model systems. METHODS: We implanted immunologically "cold" murine PDA cells orthotopically in wide type C57BL/6J mice. We administered combinations of inhibitors of MEK1/2, inhibitors of autophagy, and aCD40 and measured anticancer efficacy and immune sequelae using mass cytometry and multiplexed immunofluorescence imaging analysis to characterize the tumor microenvironment. We also used human and mouse PDA cell lines and human macrophages in vitro to perform functional assays to elucidate the cellular effects induced by the treatments. RESULTS: We find that coinhibition of MEK (using cobimetinib) and autophagy (using mefloquine), but not either treatment alone, activates the STING/type I interferon pathway in tumor cells that in turn activates paracrine tumor associated macrophages toward an immunogenic M1-like phenotype. This switch is further augmented by aCD40. Triple therapy (cobimetinib + mefloquine + aCD40) achieved cytotoxic T-cell activation in an immunologically "cold" mouse PDA model, leading to enhanced antitumor immunity. CONCLUSIONS: MEK and autophagy coinhibition coupled with aCD40 invokes immune repolarization and is an attractive therapeutic approach for PDA immunotherapy development.


Asunto(s)
Autofagia/inmunología , Azetidinas/farmacología , Antígenos CD40/agonistas , Carcinoma Ductal Pancreático/inmunología , Mefloquina/farmacología , Neoplasias Pancreáticas/inmunología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Hidroxicloroquina/farmacología , Inmunoterapia , Interferón Tipo I/efectos de los fármacos , Interferón Tipo I/inmunología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Macrófagos , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/inmunología , Ratones , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/inmunología , Escape del Tumor , Microambiente Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/efectos de los fármacos
4.
Nat Commun ; 12(1): 6798, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815397

RESUMEN

G-protein-coupled receptors (GPCRs), especially chemokine receptors, play a central role in the regulation of T cell migration. Various GPCRs are upregulated in activated CD4 T cells, including P2Y10, a putative lysophospholipid receptor that is officially still considered an orphan GPCR, i.e., a receptor with unknown endogenous ligand. Here we show that in mice lacking P2Y10 in the CD4 T cell compartment, the severity of experimental autoimmune encephalomyelitis and cutaneous contact hypersensitivity is reduced. P2Y10-deficient CD4 T cells show normal activation, proliferation and differentiation, but reduced chemokine-induced migration, polarization, and RhoA activation upon in vitro stimulation. Mechanistically, CD4 T cells release the putative P2Y10 ligands lysophosphatidylserine and ATP upon chemokine exposure, and these mediators induce P2Y10-dependent RhoA activation in an autocrine/paracrine fashion. ATP degradation impairs RhoA activation and migration in control CD4 T cells, but not in P2Y10-deficient CD4 T cells. Importantly, the P2Y10 pathway appears to be conserved in human T cells. Taken together, P2Y10 mediates RhoA activation in CD4 T cells in response to auto-/paracrine-acting mediators such as LysoPS and ATP, thereby facilitating chemokine-induced migration and, consecutively, T cell-mediated diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Receptores Purinérgicos P2Y/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Animales , Comunicación Autocrina/inmunología , Linfocitos T CD4-Positivos/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Quimiocinas/metabolismo , Quimiotaxis de Leucocito/inmunología , Encefalomielitis Autoinmune Experimental/sangre , Femenino , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Humanos , Lisofosfolípidos/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y/genética , Proteína de Unión al GTP rhoA/metabolismo
5.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575956

RESUMEN

Umbilical cord blood (UCB) has long been seen as a rich source of naïve cells with strong regenerative potential, likely mediated by paracrine signals. More recently, small extracellular vesicles (sEV), such as exosomes, have been shown to play essential roles in cell-to-cell communication, via the transport of numerous molecules, including small RNAs. Often explored for their potential as biomarkers, sEV are now known to have regenerative and immunomodulating characteristics, particularly if isolated from stem cell-rich tissues. In this study, we aim to characterize the immunomodulating properties of umbilical cord blood mononuclear cell-derived sEV (UCB-MNC-sEV) and explore their therapeutic potential for inflammatory skin diseases. UCB-MNC-sEV were shown to shift macrophages toward an anti-inflammatory phenotype, which in turn exert paracrine effects on fibroblasts, despite previous inflammatory stimuli. Additionally, the incubation of PBMC with UCB-MNC-sEV resulted in a reduction of total CD4+ and CD8+ T-cell proliferation and cytokine release, while specifically supporting the development of regulatory T-cells (Treg), by influencing FOXP3 expression. In a 3D model of psoriatic skin, UCB-MNC-sEV reduced the expression of inflammatory and psoriatic markers IL6, IL8, CXCL10, COX2, S100A7, and DEFB4. In vivo, UCB-MNC-sEV significantly prevented or reversed acanthosis in imiquimod-induced psoriasis, and tendentially increased the number of Treg in skin, without having an overall impact on disease burden. This work provides evidence for the anti-inflammatory and tolerogenic effect of UCB-MNC-sEV, which may be harnessed for the treatment of Th17-driven inflammatory skin diseases, such as psoriasis.


Asunto(s)
Exosomas/inmunología , Factores de Transcripción Forkhead/genética , Inmunomodulación/inmunología , Inflamación/terapia , Psoriasis/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular/genética , Comunicación Celular/inmunología , Proliferación Celular/genética , Citocinas/genética , Exosomas/genética , Exosomas/trasplante , Vesículas Extracelulares/trasplante , Femenino , Sangre Fetal/inmunología , Sangre Fetal/trasplante , Humanos , Inmunomodulación/genética , Inflamación/sangre , Inflamación/patología , Macrófagos/inmunología , Masculino , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Psoriasis/sangre , Psoriasis/patología , Linfocitos T Reguladores/inmunología
6.
Adv Protein Chem Struct Biol ; 126: 39-62, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34090619

RESUMEN

Mesenchymal stem cells (MSC) are adult stem cells which reside in almost all postnatal tissue where, in juxtacrine and paracrine manner, regulate phenotype and function of immune cells, maintain tissue homeostasis, attenuate on-going inflammation and promote repair and regeneration of injured tissues. Due to their capacity to suppress detrimental immune response, MSC have been considered as potentially new therapeutic agents in the treatment of autoimmune and inflammatory diseases. It was recently revealed that apoptosis may increase anti-inflammatory properties of MSC by enhancing their capacity to induce generation of immunosuppressive phenotype in macrophages and dendritic cells. Upon phagocytosis, apoptotic MSC induce generation of immunosuppressive phenotype in monocytes/macrophages and promote production of anti-inflammatory cytokines and growth factors that attenuate inflammation and facilitate repair and regeneration of injured tissues. Importantly, immunomodulation mediated by apoptotic MSC was either similar or even better than immunomodulation accomplished by viable MSC. In contrast to viable MSC, which obtain either pro- or anti-inflammatory phenotype upon engraftment in different tissue microenvironments, apoptotic MSC were not subject to changes in their immunomodulatory characteristics upon diverse stimuli, indicating their potential for clinical use. In this chapter, we summarized current knowledge about beneficial effects of apoptotic MSC in the suppression of detrimental local and systemic immune response, and we emphasized their therapeutic potential in the treatment of inflammatory diseases.


Asunto(s)
Apoptosis/inmunología , Enfermedades Autoinmunes/inmunología , Tolerancia Inmunológica , Células Madre Mesenquimatosas/inmunología , Animales , Enfermedades Autoinmunes/terapia , Humanos , Inflamación/inmunología , Inflamación/terapia , Macrófagos/inmunología , Monocitos/inmunología , Comunicación Paracrina/inmunología , Fagocitosis
7.
J Reprod Immunol ; 146: 103330, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34049032

RESUMEN

During pregnancy, the semi-allogeneic nature of the foetus requires maternal immune adaption and acquisition of tolerance at the foetal-maternal interface. Macrophages with regulatory properties and regulatory T (Treg) cells are central in promoting foetal tolerance and are enriched in the decidua (the uterine endometrium during pregnancy). Although tissue-resident decidual stromal cells (DSC) have been implicated in regulatory functions, it is not known if they are able to induce the regulatory phenotype of macrophages and T-cells. In this study we report that maternally derived DSC are able to induce homeostatic M2 macrophages and Treg cells. CD14+ monocytes and CD4+ T-cells from healthy non-pregnant women were cultured in the presence or absence of conditioned medium (CM) from DSC isolated from 1st trimester and term placentas. DSC-CM alone was able to promote the survival of macrophages and to induce a regulatory CD14brightCD163+CD209+CD86dim phenotype, typical for decidual macrophages and similar to that induced by M-CSF. Interestingly, DSC-CM was also able to overrule the pro-inflammatory effects of GM-CSF by upregulating CD14, CD163 and CD209. Protein-profiling showed that M-CSF was secreted by DSC, and blocking of M-CSF partially reversed the M2 phenotype and reduced viability. DSC-CM also expanded CD25brightFoxp3+ Treg cells, an expansion that was abolished by a SMAD3-inhibitor, indicating the contribution of TGF-ß signaling. In conclusion, our findings collectively emphasize the role of tissue-resident stromal cells in shaping the tolerogenic environment at the foetal-maternal interface.


Asunto(s)
Decidua/inmunología , Tolerancia Inmunológica , Macrófagos/inmunología , Células del Estroma/inmunología , Linfocitos T Reguladores/inmunología , Aborto Inducido , Adulto , Supervivencia Celular/inmunología , Células Cultivadas , Cesárea , Medios de Cultivo Condicionados/metabolismo , Decidua/citología , Decidua/metabolismo , Femenino , Humanos , Intercambio Materno-Fetal/inmunología , Comunicación Paracrina/inmunología , Embarazo , Primer Trimestre del Embarazo/inmunología , Tercer Trimestre del Embarazo/inmunología , Cultivo Primario de Células , Células del Estroma/metabolismo
8.
Circulation ; 143(22): 2169-2187, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33726497

RESUMEN

BACKGROUND: Arrhythmogenic cardiomyopathy (ACM) manifests with sudden death, arrhythmias, heart failure, apoptosis, and myocardial fibro-adipogenesis. The phenotype typically starts at the epicardium and advances transmurally. Mutations in genes encoding desmosome proteins, including DSP (desmoplakin), are major causes of ACM. METHODS: To delineate contributions of the epicardium to the pathogenesis of ACM, the Dsp allele was conditionally deleted in the epicardial cells in mice upon expression of tamoxifen-inducible Cre from the Wt1 locus. Wild type (WT) and Wt1-CreERT2:DspW/F were crossed to Rosa26mT/mG (R26mT/mG) dual reporter mice to tag the epicardial-derived cells with the EGFP (enhanced green fluorescent protein) reporter protein. Tagged epicardial-derived cells from adult Wt1-CreERT2:R26mT/mG and Wt1-CreERT2: R26mT/mG:DspW/F mouse hearts were isolated by fluorescence-activated cell staining and sequenced by single-cell RNA sequencing. RESULTS: WT1 (Wilms tumor 1) expression was progressively restricted postnatally and was exclusive to the epicardium by postnatal day 21. Expression of Dsp was reduced in the epicardial cells but not in cardiac myocytes in the Wt1-CreERT2:DspW/F mice. The Wt1-CreERT2:DspW/F mice exhibited premature death, cardiac dysfunction, arrhythmias, myocardial fibro-adipogenesis, and apoptosis. Single-cell RNA sequencing of ≈18 000 EGFP-tagged epicardial-derived cells identified genotype-independent clusters of endothelial cells, fibroblasts, epithelial cells, and a very small cluster of cardiac myocytes, which were confirmed on coimmunofluorescence staining of the myocardial sections. Differentially expressed genes between the paired clusters in the 2 genotypes predicted activation of the inflammatory and mitotic pathways-including the TGFß1 (transforming growth factor ß1) and fibroblast growth factors-in the epicardial-derived fibroblast and epithelial clusters, but predicted their suppression in the endothelial cell cluster. The findings were corroborated by analysis of gene expression in the pooled RNA-sequencing data, which identified predominant dysregulation of genes involved in epithelial-mesenchymal transition, and dysregulation of 146 genes encoding the secreted proteins (secretome), including genes in the TGFß1 pathway. Activation of the TGFß1 and its colocalization with fibrosis in the Wt1-CreERT2:R26mT/mG:DspW/F mouse heart was validated by complementary methods. CONCLUSIONS: Epicardial-derived cardiac fibroblasts and epithelial cells express paracrine factors, including TGFß1 and fibroblast growth factors, which mediate epithelial-mesenchymal transition, and contribute to the pathogenesis of myocardial fibrosis, apoptosis, arrhythmias, and cardiac dysfunction in a mouse model of ACM. The findings uncover contributions of the epicardial-derived cells to the pathogenesis of ACM.


Asunto(s)
Cardiomiopatías/fisiopatología , Comunicación Paracrina/inmunología , Pericardio/fisiopatología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Cardiomiopatías/mortalidad , Modelos Animales de Enfermedad , Humanos , Ratones , Análisis de Supervivencia
9.
Cell Immunol ; 363: 104316, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33713902

RESUMEN

Clinical and experimental studies have described eosinophil infiltration in Leishmania amazonensis infection sites, positioning eosinophils strategically adjacent to the protozoan-infected macrophages in cutaneous leishmaniasis. Here, by co-culturing mouse eosinophils with L. amazonensis-infected macrophages, we studied the impact of eosinophils on macrophage ability to regulate intracellular L. amazonensis infection. Eosinophils prevented the increase in amastigote numbers within macrophages by a mechanism dependent on a paracrine activity mediated by eosinophil-derived prostaglandin (PG) D2 acting on DP2 receptors. Exogenous PGD2 mimicked eosinophil-mediated effect on managing L. amazonensis intracellular infection by macrophages and therefore may function as a complementary tool for therapeutic intervention in L. amazonensis-driven cutaneous leishmaniasis.


Asunto(s)
Eosinófilos/inmunología , Leishmaniasis/inmunología , Macrófagos/inmunología , Prostaglandina D2/inmunología , Animales , Eosinófilos/metabolismo , Femenino , Leishmania/inmunología , Leishmaniasis/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Comunicación Paracrina/inmunología , Prostaglandina D2/metabolismo , Receptores de Prostaglandina/metabolismo
11.
Cell Mol Gastroenterol Hepatol ; 11(1): 249-272, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32827707

RESUMEN

BACKGROUND & AIMS: TNFSF15 genetic variants leading to increased TNF superfamily member 15 (TNFSF15) expression confer risk for inflammatory bowel disease (IBD), and TNFSF15 is being explored as a therapeutic target in IBD patients. Although the focus for TNFSF15-mediated inflammatory outcomes has been predominantly on its action on T cells, TNFSF15 also promotes inflammatory outcomes in human macrophages. Given the critical role for macrophages in bacterial clearance, we hypothesized that TNFSF15 promotes antimicrobial pathways in human macrophages and that macrophages from TNFSF15 IBD risk carriers with higher TNFSF15 expression have an advantage in these antimicrobial outcomes. METHODS: We analyzed protein expression, signaling, bacterial uptake, and intracellular bacterial clearance in human monocyte-derived macrophages through flow cytometry, enzyme-linked immunosorbent assay, and gentamicin protection. RESULTS: Autocrine/paracrine TNFSF15 interactions with death receptor 3 (DR3) were required for optimal levels of pattern-recognition-receptor (PRR)-induced bacterial clearance in human macrophages. TNFSF15 induced pyruvate dehydrogenase kinase 1-dependent bacterial uptake and promoted intracellular bacterial clearance through reactive oxygen species, nitric oxide synthase 2, and autophagy up-regulation. The TNFSF15-initiated TNF receptor-associated factor 2/receptor-interacting protein kinase 1/RIP3 pathway was required for mitogen-activated protein kinase and nuclear factor-κB activation, and, in turn, induction of each of the antimicrobial pathways; the TNFSF15-initiated Fas-associated protein with death domain/mucosa-associated lymphoid tissue lymphoma translocation protein 1/caspase-8 pathway played a less prominent role in antimicrobial functions, despite its key role in TNFSF15-induced cytokine secretion. Complementation of signaling pathways or antimicrobial pathways restored bacterial uptake and clearance in PRR-stimulated macrophages where TNFSF15:DR3 interactions were inhibited. Monocyte-derived macrophages from high TNFSF15-expressing rs6478108 TT IBD risk carriers in the TNFSF15 region showed increased levels of the identified antimicrobial pathways. CONCLUSIONS: We identify that autocrine/paracrine TNFSF15 is required for optimal PRR-enhanced antimicrobial pathways in macrophages, define mechanisms regulating TNFSF15-dependent bacterial clearance, and determine how the TNFSF15 IBD risk genotype modulates these outcomes.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Macrófagos/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Comunicación Autocrina/inmunología , Células Cultivadas , Enterococcus faecalis/inmunología , Enterococcus faecalis/aislamiento & purificación , Escherichia coli/inmunología , Escherichia coli/aislamiento & purificación , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/microbiología , Macrófagos/metabolismo , Ratones , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
12.
Plast Reconstr Surg ; 147(2): 231e-239e, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33235042

RESUMEN

BACKGROUND: Pathologic scarring including keloid and hypertrophic scar causes aesthetic and physical problems, and there are clinical difficulties (e.g., posttreatment recurrence) in dealing with pathologic scarring. Understanding the mechanisms that underlie scar control in wound healing will help prevent and treat pathologic scarring. The authors focused on CD206+ macrophages in the wound-healing process, and hypothesized that CD206+ macrophages have antifibrotic effects on fibroblasts. METHODS: The authors established a co-culture system for CD206+ macrophages and fibroblasts (cell ratio, 1:1). The authors examined the CD206+ macrophages' antifibrotic effects on fibroblasts after a 72-hour culture, focusing on fibrosis-related genes. To identify key factor(s) in the interaction between CD206+ macrophages and fibroblasts, the authors analyzed cytokines in a conditioned medium of the co-culture system. RESULTS: Under co-culture with CD206+ macrophages, expression of the following in the fibroblasts was significantly down-regulated: type 1 (fold change, 0.38) and type 3 collagen (0.45), alpha smooth muscle actin (0.24), connective tissue growth factor (0.40), and transforming growth factor-beta (0.66); the expression of matrix metalloproteinase 1 was significantly up-regulated (1.92). Conditioned medium in the co-culture showed a high interleukin (IL)-6 concentration (419 ± 88 pg/ml). When IL-6 was added to fibroblasts, antifibrotic changes in gene expression (as observed under the co-culture) occurred in the fibroblasts. CONCLUSIONS: The authors' in vitro results revealed that CD206+ macrophages have antifibrotic effects on fibroblasts by means of a paracrine mechanism involving IL-6. Understanding these effects, especially in vivo, will help elucidate the mechanism of scar control in wound healing and contribute to the development of new scar treatments.


Asunto(s)
Cicatriz Hipertrófica/inmunología , Fibroblastos/patología , Interleucina-6/metabolismo , Queloide/inmunología , Macrófagos/inmunología , Herida Quirúrgica/complicaciones , Células Cultivadas , Cicatriz Hipertrófica/patología , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Fibroblastos/inmunología , Voluntarios Sanos , Humanos , Queloide/patología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Receptores Inmunológicos/metabolismo , Piel/citología , Piel/inmunología , Piel/patología , Herida Quirúrgica/inmunología , Cicatrización de Heridas/inmunología
13.
Biochim Biophys Acta Mol Basis Dis ; 1867(2): 166027, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33248276

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease associated with dysregulated interleukin (IL)-6 and autophagy. Although such disturbances are increasingly recognized in patients with SLE and animal models of the disease, little is known about the specific role of IL-6 and autophagy in SLE macrophages. Here, we investigated alterations in the IL-6 axis and autophagy in macrophages derived from patients with SLE and determined whether IL-6 modulates autophagy using human macrophage models. Serum IL-6 detected by ELISA was higher in SLE patients (n = 19) than in normal controls (n = 19, p < 0.001). Levels of the IL-6 receptor (IL-6R) and autophagic markers LC3B and p62 in SLE and normal macrophages were assessed by real-time PCR, western blotting, and immunofluorescence. Compared with normal macrophages, SLE macrophages not only overexpressed IL-6Rs but also exhibited impaired autophagic degradation as evidenced by elevated levels of LC3B and p62. In vitro analyses using macrophage models revealed that prolonged exposure to exogenous recombinant human IL-6 induced a marked impairment of autophagic degradation indicated by elevated levels of LC3B and p62 in both primary macrophages and transformed macrophages. Pretreatment with tocilizumab, a humanized anti-IL-6R monoclonal antibody, restored autophagic degradation and reversed p62 accumulation in a paracrine manner in macrophages. These findings demonstrate that SLE involves IL-6-induced impairment of autophagic degradation through augmentation of IL-6R in human macrophages.


Asunto(s)
Autofagia/inmunología , Interleucina-6/metabolismo , Lupus Eritematoso Sistémico/inmunología , Macrófagos/metabolismo , Receptores de Interleucina-6/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Autofagia/efectos de los fármacos , Estudios de Casos y Controles , Células Cultivadas , Voluntarios Sanos , Humanos , Interleucina-6/sangre , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Macrófagos/inmunología , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/metabolismo , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/metabolismo , Receptores de Interleucina-6/análisis , Receptores de Interleucina-6/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Índice de Severidad de la Enfermedad , Células THP-1
14.
Front Immunol ; 11: 584297, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329556

RESUMEN

Multiple sclerosis (MS) is caused by a combination of genetic and environmental factors. It is believed that previous infection with Epstein Barr Virus (EBV) plays an important role in the development of MS. Previously, we developed a murine model where latent infection with gamma herpesvirus 68 (γHV-68), a murine homolog to EBV, enhanced the symptoms of experimental autoimmune encephalomyelitis (EAE), resulting in disease that more closely resembles MS in humans. Here, we explored the conditions that were necessary for EAE enhancement. We showed that latently infected CD19+IgD- B cells were capable of enhancing EAE symptoms when transferred from mice previously infected with γHV-68 into uninfected mice. We also observed a prevention of enhancement when B cells were depleted before infection. However, depletion after the establishment of latency only partially reduced EAE. This indicated the existence of a mechanism where B cells play an important role as antigen presenting cells (APCs) prior to EAE induction for the priming of Th1 cells. It is possible that these signals persist even after B cell depletion, strongly suggesting a paracrine signaling modulation of non-B cell APCs. These results strongly support the concept that EBV contributes to the development of autoimmunity and highlights the need for a vaccine against EBV that could limit or prevent multiple sclerosis development.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Animales , Células Presentadoras de Antígenos/inmunología , Autoinmunidad/inmunología , Encéfalo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/virología , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina/inmunología , Transducción de Señal/inmunología , Células TH1/inmunología
15.
Nat Commun ; 11(1): 6298, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293516

RESUMEN

Immunosuppressive tumor microenvironment (TME) and ascites-derived spheroids in ovarian cancer (OC) facilitate tumor growth and progression, and also pose major obstacles for cancer therapy. The molecular pathways involved in the OC-TME interactions, how the crosstalk impinges on OC aggression and chemoresistance are not well-characterized. Here, we demonstrate that tumor-derived UBR5, an E3 ligase overexpressed in human OC associated with poor prognosis, is essential for OC progression principally by promoting tumor-associated macrophage recruitment and activation via key chemokines and cytokines. UBR5 is also required to sustain cell-intrinsic ß-catenin-mediated signaling to promote cellular adhesion/colonization and organoid formation by controlling the p53 protein level. OC-specific targeting of UBR5 strongly augments the survival benefit of conventional chemotherapy and immunotherapies. This work provides mechanistic insights into the novel oncogene-like functions of UBR5 in regulating the OC-TME crosstalk and suggests that UBR5 is a potential therapeutic target in OC treatment for modulating the TME and cancer stemness.


Asunto(s)
Carcinoma Epitelial de Ovario/inmunología , Macrófagos Peritoneales/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Peritoneales/inmunología , Escape del Tumor/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Anciano , Animales , Ascitis/genética , Ascitis/inmunología , Ascitis/patología , Carcinoma Epitelial de Ovario/mortalidad , Carcinoma Epitelial de Ovario/secundario , Carcinoma Epitelial de Ovario/terapia , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva/métodos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Comunicación Paracrina/inmunología , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/secundario , Cultivo Primario de Células , Pronóstico , Receptores Quiméricos de Antígenos/inmunología , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ubiquitina-Proteína Ligasas/genética
16.
Biol Pharm Bull ; 43(11): 1785-1791, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33132324

RESUMEN

Mesenchymal stem cells (MSCs) are capable of repairing skeletal muscle via paracrine mechanisms. This regenerative effect of MSCs on skeletal muscle is based on promoting the proliferation and differentiation of myogenic cells and inhibiting the inflammatory response of immune cells. However, it is unclear whether MSCs affect the inflammatory response of skeletal muscle cells. In this study, we evaluated the paracrine effect of mouse MSCs on the inflammatory response of lipopolysaccharide (LPS)-stimulated C2C12 mouse myoblasts. Interleukin (IL)-6 production from LPS-stimulated C2C12 cells was significantly increased by coculture with MSCs or culture in conditioned medium of MSCs. This increased IL-6 production from C2C12 cells was not significantly suppressed by inhibiting mitogen-activated protein kinase pathways, but it was significantly suppressed by pretreatment with nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) inhibitors. In addition, IL-6 and inducible nitric oxide synthase (iNOS) mRNA expression was increased significantly in C2C12 cells cocultured with MSCs, while tumor necrosis factor (TNF)-α and IL-1ß mRNA expression was decreased. Furthermore, conditioned medium of C2C12 cells cocultured with MSCs exerted remarkable anti-inflammatory effects on LPS-stimulated mouse macrophages.


Asunto(s)
Sistema de Señalización de MAP Quinasas/inmunología , Células Madre Mesenquimatosas/metabolismo , Mioblastos Esqueléticos/inmunología , Comunicación Paracrina/inmunología , Animales , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Mioblastos Esqueléticos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Autoimmun ; 115: 102540, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32893081

RESUMEN

Lupus nephritis (LN) is a major cause for overall morbidity and mortality in patients with systemic lupus erythematosus (SLE), while its pathogenic mechanisms are still not well understood. Extracellular vesicles (EVs) are membrane vesicles that are released from almost all cell types. EVs can be subdivided into exosomes, microvesicles, and apoptotic bodies. Latest studies have shown that EVs can be released during several cellular events, including cell activation, autophagy, and several types of programed cell death, i.e. apoptosis, necroptosis, pyroptosis, and NETosis. Emerging evidence demonstrates that EVs harbor different bioactive molecules, including nucleic acids, proteins, lipids, cytokines, immune complexes (ICs), complements, and other molecules, some of which may contribute to pathogenesis of autoimmune diseases. EVs can serve as novel information shuttle to mediate local autocrine or paracrine signals to nearby cells, and distant endocrine signals to cells located far away. In LN, EVs may have pathogenic effects by transportation of autoantigens or complements, promotion of IC deposition or complement activation, and stimulation of inflammatory responses, renal tissue injury, or microthrombus formation. Additionally, EVs released from kidney cells may serve as specific biomarkers for diagnosis or monitoring of disease activity and therapeutic efficacy. In this review, we will summarize the latest progress about EV generation from basic research, their potential pathologic effects on LN, and their clinical implications. The cutting-edge knowledge about EV research provides insights into novel therapeutic strategy, new tools for diagnosis or prognosis, and evaluation approaches for treatment effectiveness in LN.


Asunto(s)
Vesículas Extracelulares/inmunología , Riñón/patología , Nefritis Lúpica/inmunología , Comunicación Autocrina/inmunología , Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Riñón/citología , Riñón/inmunología , Nefritis Lúpica/sangre , Nefritis Lúpica/diagnóstico , Nefritis Lúpica/patología , Comunicación Paracrina/inmunología , Pronóstico , Índice de Severidad de la Enfermedad
18.
EMBO J ; 39(16): e105057, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32643835

RESUMEN

Alveolar macrophages (AMs) and epithelial cells (ECs) are the lone resident lung cells positioned to respond to pathogens at early stages of infection. Extracellular vesicles (EVs) are important vectors of paracrine signaling implicated in a range of (patho)physiologic contexts. Here we demonstrate that AMs, but not ECs, constitutively secrete paracrine activity localized to EVs which inhibits influenza infection of ECs in vitro and in vivo. AMs exposed to cigarette smoke extract lost the inhibitory activity of their secreted EVs. Influenza strains varied in their susceptibility to inhibition by AM-EVs. Only those exhibiting early endosomal escape and high pH of fusion were inhibited via a reduction in endosomal pH. By contrast, strains exhibiting later endosomal escape and lower fusion pH proved resistant to inhibition. These results extend our understanding of how resident AMs participate in host defense and have broader implications in the defense and treatment of pathogens internalized within endosomes.


Asunto(s)
Endosomas , Vesículas Extracelulares/inmunología , Virus de la Influenza A/inmunología , Macrófagos Alveolares/inmunología , Comunicación Paracrina/inmunología , Internalización del Virus , Células A549 , Animales , Perros , Endosomas/inmunología , Endosomas/patología , Endosomas/virología , Células HEK293 , Humanos , Macrófagos Alveolares/patología , Células de Riñón Canino Madin Darby , Ratones , Ratas , Ratas Wistar , Células THP-1
19.
Nat Rev Cancer ; 20(8): 455-469, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32546840

RESUMEN

Prostate cancer is a major cause of cancer morbidity and mortality. Intra-prostatic inflammation is a risk factor for prostate carcinogenesis, with diet, chemical injury and an altered microbiome being causally implicated. Intra-prostatic inflammatory cell recruitment and expansion can ultimately promote DNA double-strand breaks and androgen receptor activation in prostate epithelial cells. The activation of the senescence-associated secretory phenotype fuels further 'inflammatory storms', with free radicals leading to further DNA damage. This drives the overexpression of DNA repair and tumour suppressor genes, rendering these genes susceptible to mutagenic insults, with carcinogenesis accelerated by germline DNA repair gene defects. We provide updates on recent advances in elucidating prostate carcinogenesis and explore novel therapeutic and prevention strategies harnessing these discoveries.


Asunto(s)
Carcinogénesis/inmunología , Inflamación/inmunología , Próstata/inmunología , Neoplasias de la Próstata/inmunología , Receptores Androgénicos/inmunología , Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Enfermedad Crónica , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Daño del ADN/inmunología , Reparación del ADN/genética , Reparación del ADN/inmunología , Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/inmunología , Humanos , Inflamación/etiología , Inflamación/genética , Masculino , Microbiota/inmunología , Obesidad/complicaciones , Obesidad/inmunología , Comunicación Paracrina/inmunología , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
20.
Integr Biol (Camb) ; 12(7): 175-187, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32556172

RESUMEN

Hematopoietic stem cells (HSCs) primarily reside in the bone marrow, where they receive external cues from their local microenvironment. The complex milieu of biophysical cues, cellular components and cell-secreted factors regulates the process by which HSC produce the blood and immune system. We previously showed direct coculture of primary murine hematopoietic stem and progenitor cells with a population of marrow-derived mesenchymal stromal and progenitor cells (MSPCs) in a methacrylamide-functionalized gelatin (GelMA) hydrogel improves hematopoietic progenitor maintenance. However, the mechanism by which MSPCs influenced HSC fate decisions remained unknown. Herein, we report the use of proteomic analysis to correlate HSC phenotype to a broad candidate pool of 200 soluble factors produced by combined mesenchymal and hematopoietic progeny. Partial least squares regression (PLSR), along with an iterative filter method, identified TGFß-1, MMP-3, c-RP and TROY as positively correlated with HSC maintenance. Experimentally, we then observe exogenous stimulation of HSC monocultures in GelMA hydrogels with these combined cytokines increases the ratio of hematopoietic progenitors to committed progeny after a 7-day culture 7.52 ± 3.65-fold compared to non-stimulated monocultures. Findings suggest a cocktail of the downselected cytokines amplifies hematopoietic maintenance potential of HSCs beyond that of MSPC-secreted factors alone. This work integrates empirical and computation methods to identify cytokine combinations to improve HSC maintenance within an engineered HSC niche, suggesting a route toward identifying feeder-free culture platforms for HSC expansion. Insight Hematopoietic stem cells within an artificial niche receive maintenance cues in the form of soluble factors from hematopoietic and mesenchymal progeny. Applying a proteomic regression analysis, we identify a reduced set of soluble factors correlated to maintenance of a hematopoietic phenotype during culture in a biomaterial model of the bone marrow niche. We identify a minimum factor cocktail that promotes hematopoietic maintenance potential in a gelatin-based culture, regardless of the presence of mesenchymal feeder cells. By combining empirical and computational methods, we report an experimentally feasible number of factors from a large dataset, enabling exogenous integration of soluble factors into an engineered hematopoietic stem cell for enhanced maintenance potential of a quiescent stem cell population.


Asunto(s)
Médula Ósea/metabolismo , Ingeniería Celular , Células Madre Hematopoyéticas/metabolismo , Nicho de Células Madre/genética , Algoritmos , Animales , Comunicación Autocrina/inmunología , Células Cultivadas , Citocinas/metabolismo , Femenino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Comunicación Paracrina/inmunología , Fenotipo , Proteómica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA