Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.513
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 632(8025): 637-646, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39085603

RESUMEN

Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.


Asunto(s)
Inmunoglobulina A , Tejido Linfoide , Mucosa Nasal , Células Plasmáticas , Linfocitos T , Cornetes Nasales , Animales , Femenino , Masculino , Ratones , Bacterias/inmunología , Movimiento Celular , Quimiocinas CC/inmunología , Quimiocinas CC/metabolismo , Centro Germinal/inmunología , Centro Germinal/citología , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/citología , Ratones Endogámicos C57BL , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Linfocitos T/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo , Cornetes Nasales/citología , Cornetes Nasales/inmunología , Vacunación , Administración Intranasal , Vacunas/inmunología , Simbiosis
2.
J Immunol ; 211(1): 118-129, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37222480

RESUMEN

Four major mucosal-associated chemokines, CCL25, CCL28, CXCL14, and CXCL17, play an important role in protecting mucosal surfaces from infectious pathogens. However, their role in protection against genital herpes remains to be fully explored. The CCL28 is a chemoattractant for the CCR10 receptor-expressing immune cells and is produced homeostatically in the human vaginal mucosa (VM). In this study, we investigated the role of the CCL28/CCR10 chemokine axis in mobilizing protective antiviral B and T cell subsets into the VM site of herpes infection. We report a significant increase in the frequencies of HSV-specific memory CCR10+CD44+CD8+ T cells, expressing high levels of CCR10, in herpes-infected asymptomatic (ASYMP) women compared with symptomatic women. Similarly, a significant increase in the CCL28 chemokine (a ligand of CCR10), was detected in the VM of herpes-infected ASYMP C57BL/6 mice, associated with the mobilization of high frequencies of HSV-specific effector memory CCR10+CD44+CD62L-CD8+ TEM cells and memory CCR10+B220+CD27+ B cells in the VM of HSV-infected ASYMP mice. Inversely, compared with wild-type C57BL/6 mice, the CCL28 knockout (CCL28-/-) mice (1) appeared to be more susceptible to intravaginal infection and reinfection with HSV type 2, and (2) exhibited a significant decrease in the frequencies of HSV-specific effector memory CCR10+CD44+CD62L-CD8+ TEM cells and of memory CD27+B220+ B cells in the infected VM. These findings suggest a critical role of the CCL28/CCR10 chemokine axis in the mobilization of antiviral memory B and T cells within the VM to protect against genital herpes infection and disease.


Asunto(s)
Herpes Genital , Humanos , Femenino , Ratones , Animales , Antivirales/metabolismo , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos , Herpesvirus Humano 2 , Membrana Mucosa , Factores de Restricción Antivirales , Receptores CCR10/metabolismo , Quimiocinas CC/metabolismo , Receptores de Hialuranos/metabolismo
3.
Biochemistry ; 63(18): 2235-2239, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39194151

RESUMEN

The poxvirus-derived protein vCCI (viral CC chemokine inhibitor) binds almost all members of the CC chemokine family with nanomolar affinity, inhibiting their pro-inflammatory actions. Understanding the affinity and specificity of vCCI could lead to new anti-inflammatory therapeutics. CCL17, also known as TARC, is unusual among CC chemokines by having only micromolar binding to vCCI. We have used sequence analysis and molecular simulations to determine the cause of this weak binding, which identified several locations in CCL17 where mutations seemed likely to improve binding to vCCI. Based on the aforementioned analysis, we expressed and tested multiple mutants of CCL17. We found two single point mutants V44K and Q45R that increased binding affinity to vCCI by 2-3-fold and, in combination, further improved affinity by 7-fold. The CCL17 triple mutant G17R/V44K/Q45R yielded a Kd of 0.25 ± 0.13 µM, a 68-fold improvement in affinity compared to the complex with wild-type CCL17. A quadruple mutant G17R/V44K/Q45R/R57W showed high affinity (0.59 ± 0.09 µM) compared to the wild type but lower affinity than the triple mutant. This work demonstrates that sequence comparisons and molecular simulations can predict chemokine mutations that increase the level of binding to vCCI, an important first step in developing engineered chemokine inhibitors useful for anti-inflammatory therapy.


Asunto(s)
Quimiocina CCL17 , Unión Proteica , Proteínas Virales , Quimiocina CCL17/metabolismo , Quimiocina CCL17/química , Quimiocina CCL17/genética , Humanos , Proteínas Virales/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Quimiocinas CC/metabolismo , Quimiocinas CC/química , Quimiocinas CC/genética , Mutación
4.
Cancer Sci ; 115(3): 777-790, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38228495

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a highly malignant and aggressive cancer whose incidence and mortality continue to increase, whereas its prognosis remains dismal. Tumor-associated macrophages (TAMs) promote malignant progression and immune microenvironment remodeling through direct contact and secreted mediators. Targeting TAMs has emerged as a promising strategy for ICC treatment. Here, we revealed the potential regulatory function of immune responsive gene 1 (IRG1) in macrophage polarization. We found that IRG1 expression remained at a low level in M2 macrophages. IRG1 overexpression can restrain macrophages from polarizing to the M2 type, which results in inhibition of the proliferation, invasion, and migration of ICC, whereas IRG1 knockdown exerts the opposite effects. Mechanistically, IRG1 inhibited the tumor-promoting chemokine CCL18 and thus suppressed ICC progression by regulating STAT3 phosphorylation. The intervention of IRG1 expression in TAMs may serve as a potential therapeutic target for delaying ICC progression.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Colangiocarcinoma/patología , Macrófagos/metabolismo , Pronóstico , Conductos Biliares Intrahepáticos/metabolismo , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Microambiente Tumoral , Quimiocinas CC/metabolismo , Factor de Transcripción STAT3/metabolismo
5.
BMC Cancer ; 24(1): 827, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992592

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths globally, influenced by aberrant circRNA expression. Investigating circRNA-miRNA-mRNA interactions can unveil underlying mechanisms of HCC and identify potential therapeutic targets. METHODS: In this study, we conducted differential analyses of mRNAs, miRNAs, and circRNAs, and established their relationships using various databases such as miRanda, miRDB, and miTarBase. Additionally, functional enrichment and immune infiltration analyses were performed to evaluate the roles of key genes. We also conducted qPCR assays and western blotting (WB) to examine the expression levels of circRNA, CCL25, and MAP2K1 in both HCC cells and clinical samples. Furthermore, we utilized overexpression and knockdown techniques for circ_0000069 and conducted wound healing, transwell invasion assays, and a tumorigenesis experiment to assess the migratory and invasive abilities of HCC cells. RESULTS: Our findings revealed significant differential expression of 612 upregulated genes and 1173 downregulated genes in HCC samples compared to normal liver tissue. Additionally, 429 upregulated circRNAs and 453 downregulated circRNAs were identified. Significantly, circ_0000069 exhibited upregulation in HCC tissues and cell lines. The overexpression of circ_0000069 notably increased the invasion and migration capacity of Huh7 cells, whereas the downregulation of circ_0000069 reduced this capability in HepG2 cells. Furthermore, this effect was counteracted by CCL25 silencing or overexpression, separately. Animal studies further confirmed that the overexpression of hsa_circ_0000069 facilitated tumor growth in xenografted nude mice, while the inhibition of CCL25 attenuated this effect. CONCLUSION: Circ_0000069 appears to promote HCC progression by regulating CCL25, suggesting that both circ_0000069 and CCL25 can serve as potential therapeutic targets.


Asunto(s)
Carcinoma Hepatocelular , Quimiocinas CC , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas , ARN Circular , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , ARN Circular/genética , Animales , Ratones , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Ratones Desnudos , MicroARNs/genética , Proliferación Celular/genética , Masculino
6.
Biopolymers ; 115(2): e23557, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37341434

RESUMEN

Chemokines are important immune system proteins, many of which mediate inflammation due to their function to activate and cause chemotaxis of leukocytes. An important anti-inflammatory strategy is therefore to bind and inhibit chemokines, which leads to the need for biophysical studies of chemokines as they bind various possible partners. Because a successful anti-chemokine drug should bind at low concentrations, techniques such as fluorescence anisotropy that can provide nanomolar signal detection are required. To allow fluorescence experiments to be carried out on chemokines, a method is described for the production of fluorescently labeled chemokines. First, a fusion-tagged chemokine is produced in Escherichia coli, then efficient cleavage of the N-terminal fusion partner is carried out with lab-produced enterokinase, followed by covalent modification with a fluorophore, mediated by the lab-produced sortase enzyme. This overall process reduces the need for expensive commercial enzymatic reagents. Finally, we utilize the product, vMIP-fluor, in binding studies with the chemokine binding protein vCCI, which has great potential as an anti-inflammatory therapeutic, showing a binding constant for vCCI:vMIP-fluor of 0.37 ± 0.006 nM. We also show how a single modified chemokine homolog (vMIP-fluor) can be used in competition assays with other chemokines and we report a Kd for vCCI:CCL17 of 14 µM. This work demonstrates an efficient method of production and fluorescent labeling of chemokines for study across a broad range of concentrations.


Asunto(s)
Quimiocinas CC , Enteropeptidasa , Humanos , Quimiocinas CC/química , Quimiocinas CC/metabolismo , Quimiocinas/química , Quimiocinas/metabolismo , Inflamación , Antiinflamatorios
7.
J Immunol ; 208(2): 303-320, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34930780

RESUMEN

The deficiency of Aire, a transcriptional regulator whose defect results in the development of autoimmunity, is associated with reduced expression of tissue-restricted self-Ags (TRAs) in medullary thymic epithelial cells (mTECs). Although the mechanisms underlying Aire-dependent expression of TRAs need to be explored, the physical identification of the target(s) of Aire has been hampered by the low and promiscuous expression of TRAs. We have tackled this issue by engineering mice with augmented Aire expression. Integration of the transcriptomic data from Aire-augmented and Aire-deficient mTECs revealed that a large proportion of so-called Aire-dependent genes, including those of TRAs, may not be direct transcriptional targets downstream of Aire. Rather, Aire induces TRA expression indirectly through controlling the heterogeneity of mTECs, as revealed by single-cell analyses. In contrast, Ccl25 emerged as a canonical target of Aire, and we verified this both in vitro and in vivo. Our approach has illuminated the Aire's primary targets while distinguishing them from the secondary targets.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Quimiocinas CC/metabolismo , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Autoinmunidad/genética , Quimiocinas CC/genética , Células Epiteliales/inmunología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Timo/citología , Factores de Transcripción/genética , Transcripción Genética/genética , Proteína AIRE
8.
Bioorg Chem ; 145: 107181, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38354503

RESUMEN

The human CC chemokine receptor 8 (CCR8) has been extensively pursued as target for the treatment of various inflammatory disorders. More recently, the importance of CCR8 in the tumor microenvironment has been demonstrated, spurring the interest in CCR8 antagonism as therapeutic strategy in immuno-oncology. On a previously described naphthalene sulfonamide with CCR8 antagonistic properties, the concept of isosterism was applied, leading to the discovery of novel CCR8 antagonists with IC50 values in the nM range in both the CCL1 competition binding and CCR8 calcium mobilization assay. The excellent CCR8 antagonistic activity of the most potent congeners was rationalized by homology molecular modeling.


Asunto(s)
Quimiocinas CC , Receptores de Quimiocina , Humanos , Quimiocinas CC/metabolismo , Quimiocina CCL1/metabolismo , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Amidas , Receptores CCR8 , Sulfonamidas/farmacología , Naftalenos/farmacología
9.
Int J Mol Sci ; 25(9)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38731942

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) can originate from acinar-to-ductal metaplasia (ADM). Pancreatic acini harboring oncogenic Kras mutations are transdifferentiated to a duct-like phenotype that further progresses to become pancreatic intraepithelial neoplasia (PanIN) lesions, giving rise to PDAC. Although ADM formation is frequently observed in KrasG12D transgenic mouse models of PDAC, the exact mechanisms of how oncogenic KrasG12D regulates this process remain an enigma. Herein, we revealed a new downstream target of oncogenic Kras, cytokine CCL9, during ADM formation. Higher levels of CCL9 and its receptors, CCR1 and CCR3, were detected in ADM regions of the pancreas in p48cre:KrasG12D mice and human PDAC patients. Knockdown of CCL9 in KrasG12D-expressed pancreatic acini reduced KrasG12D-induced ADM in a 3D organoid culture system. Moreover, exogenously added recombinant CCL9 and overexpression of CCL9 in primary pancreatic acini induced pancreatic ADM. We also showed that, functioning as a downstream target of KrasG12D, CCL9 promoted pancreatic ADM through upregulation of the intracellular levels of reactive oxygen species (ROS) and metalloproteinases (MMPs), including MMP14, MMP3 and MMP2. Blockade of MMPs via its generic inhibitor GM6001 or knockdown of specific MMP such as MMP14 and MMP3 decreased CCL9-induced pancreatic ADM. In p48cre:KrasG12D transgenic mice, blockade of CCL9 through its specific neutralizing antibody attenuated pancreatic ADM structures and PanIN lesion formation. Furthermore, it also diminished infiltrating macrophages and expression of MMP14, MMP3 and MMP2 in the ADM areas. Altogether, our results provide novel mechanistic insight into how oncogenic Kras enhances pancreatic ADM through its new downstream target molecule, CCL9, to initiate PDAC.


Asunto(s)
Células Acinares , Carcinoma Ductal Pancreático , Metaplasia , Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Especies Reactivas de Oxígeno , Animales , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Humanos , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Metaplasia/metabolismo , Metaplasia/genética , Células Acinares/metabolismo , Células Acinares/patología , Ratones Transgénicos , Quimiocinas CC/metabolismo , Quimiocinas CC/genética , Proteínas Inflamatorias de Macrófagos/metabolismo , Proteínas Inflamatorias de Macrófagos/genética , Páncreas/metabolismo , Páncreas/patología
10.
Br J Cancer ; 129(3): 426-443, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37380804

RESUMEN

BACKGROUND: The epigenetic mechanisms involved in the progression of pancreatic ductal adenocarcinoma (PDAC) remain largely unexplored. This study aimed to identify key transcription factors (TFs) through multiomics sequencing to investigate the molecular mechanisms of TFs that play critical roles in PDAC. METHODS: To characterise the epigenetic landscape of genetically engineered mouse models (GEMMs) of PDAC with or without KRAS and/or TP53 mutations, we employed ATAC-seq, H3K27ac ChIP-seq, and RNA-seq. The effect of Fos-like antigen 2 (FOSL2) on survival was assessed using the Kaplan-Meier method and multivariate Cox regression analysis for PDAC patients. To study the potential targets of FOSL2, we performed Cleavage Under Targets and Tagmentation (CUT&Tag). To explore the functions and underlying mechanisms of FOSL2 in PDAC progression, we employed several assays, including CCK8, transwell migration and invasion, RT-qPCR, Western blotting analysis, IHC, ChIP-qPCR, dual-luciferase reporter, and xenograft models. RESULTS: Our findings indicated that epigenetic changes played a role in immunosuppressed signalling during PDAC progression. Moreover, we identified FOSL2 as a critical regulator that was up-regulated in PDAC and associated with poor prognosis in patients. FOSL2 promoted cell proliferation, migration, and invasion. Importantly, our research revealed that FOSL2 acted as a downstream target of the KRAS/MAPK pathway and recruited regulatory T (Treg) cells by transcriptionally activating C-C motif chemokine ligand 28 (CCL28). This discovery highlighted the role of an immunosuppressed regulatory axis involving KRAS/MAPK-FOSL2-CCL28-Treg cells in the development of PDAC. CONCLUSION: Our study uncovered that KRAS-driven FOSL2 promoted PDAC progression by transcriptionally activating CCL28, revealing an immunosuppressive role for FOSL2 in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Ratones , Humanos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regulación hacia Arriba , Cromatina , Ligandos , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Proliferación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Quimiocinas CC/metabolismo , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Neoplasias Pancreáticas
11.
Am J Transplant ; 23(10): 1536-1550, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37394140

RESUMEN

The present study aims to elucidate the possible involvement of H19 in primary graft dysfunction (PGD) following lung transplantation (LT) and the underlying mechanism. The transcriptome data were obtained through high-throughput sequencing analysis, and the differential long noncoding RNAs and messenger RNAs were screened for coexpression analysis. The interaction among H19, KLF5 and CCL28 was analyzed. A hypoxia-induced human pulmonary microvascular endothelial cell injury model was established, in which H19 was knocked down to elucidate its effect on the lung function, inflammatory response, and cell apoptosis. An orthotopic left LT model was constructed for in vivo mechanistic validation. High-throughput transcriptome sequencing analysis revealed the involvement of the H19/KLF5/CCL28 signaling axis in PGD. Silencing of H19 reduced inflammatory response and thus improved PGD. CCL28 secreted by human pulmonary microvascular endothelial cells after LT recruited neutrophils and macrophages. Mechanistic investigations indicated that H19 augmented the expression of CCL28 by binding to the transcription factor KLF5. Abundant expression of CCL28 reversed the alleviating effect of H19 silencing on PGD. In conclusion, the results point out that H19 exerts a promoting effect on PGD through increasing KLF5 expression and the subsequent CCL28 expression. Our study provides a novel insight into the mechanism of action of H19.


Asunto(s)
Trasplante de Pulmón , MicroARNs , Disfunción Primaria del Injerto , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Células Endoteliales/metabolismo , Disfunción Primaria del Injerto/etiología , Regulación de la Expresión Génica , Trasplante de Pulmón/efectos adversos , MicroARNs/genética , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo
12.
Cancer Immunol Immunother ; 72(4): 903-916, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36161509

RESUMEN

Tumor-associated macrophages (TAMs) play an important role in tumor growth and metastasis. However, the involvement of TAMs infiltration in pulmonary osteosarcoma (OS) metastasis remains poorly understood. Therefore, the effect of OS cells on macrophages migration was investigated by in vivo and in vitro experiments to evaluate the infiltration and mechanism of TAMs in pulmonary OS metastases. The results showed that the zinc finger protein ZIM3 was upregulated in OS cells than in osteoblasts and activated the expression of CCL25, which subsequently promoted the migration of M2 macrophages. CCL25 or ZIM3 silencing in OS cells inhibited the infiltration of M2 macrophages and the formation of pulmonary metastatic nodules in a mouse model of pulmonary OS metastasis and prolonged the survival of the mice. Furthermore, bioinformatics analyses revealed that CCL25 and ZIM3 expressions are negatively correlated with the prognosis of OS patients. In conclusion, this study found that a large number of M2 TAMs were recruited into pulmonary metastatic nodules of OS through the activation of the ZIM3-CCL25 axis in OS cells, thereby facilitating OS metastasis. Therefore, the suppression of ZIM3-CCL25-induced recruitment of M2 TAMs to the metastatic sites might be considered as a therapeutic approach to inhibit the growth of pulmonary OS metastases.


Asunto(s)
Neoplasias Óseas , Neoplasias Pulmonares , Osteosarcoma , Animales , Ratones , Macrófagos/metabolismo , Línea Celular Tumoral , Pronóstico , Neoplasias Pulmonares/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/tratamiento farmacológico , Neoplasias Óseas/genética , Microambiente Tumoral , Quimiocinas CC/metabolismo , Quimiocinas CC/farmacología , Quimiocinas CC/uso terapéutico
13.
Nat Immunol ; 12(9): 879-87, 2011 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-21841786

RESUMEN

Little is known about the molecular mechanisms that determine the entry into the lymph node and intranodal positioning of lymph-derived cells. By injecting cells directly into afferent lymph vessels of popliteal lymph nodes, we demonstrate that lymph-derived T cells entered lymph-node parenchyma mainly from peripheral medullary sinuses, whereas dendritic cells (DCs) transmigrated through the floor of the subcapsular sinus on the afferent side. Transmigrating DCs induced local changes that allowed the concomitant entry of T cells at these sites. Signals mediated by the chemokine receptor CCR7 were absolutely required for the directional migration of both DCs and T cells into the T cell zone but were dispensable for the parenchymal entry of lymph-derived T cells and dendrite probing of DCs. Our findings provide insight into the molecular and structural requirements for the entry into lymph nodes and intranodal migration of lymph-derived cells of the immune system.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiocinas CC/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Receptores CCR7/inmunología , Migración Transcelular de la Célula/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Quimiocinas CC/metabolismo , Células Dendríticas/citología , Citometría de Flujo , Humanos , Inyecciones Intralinfáticas , Linfa/inmunología , Ganglios Linfáticos/citología , Vasos Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores CCR7/deficiencia , Receptores CCR7/genética
14.
Hepatology ; 75(3): 518-530, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34633679

RESUMEN

BACKGROUND AND AIMS: The "gut homing" hypothesis suggests the pathogenesis of primary sclerosing cholangitis (PSC) is driven by aberrant hepatic expression of gut adhesion molecules and subsequent recruitment of gut-derived T cells to the liver. However, inconsistencies lie within this theory including an absence of investigations and comparisons with other chronic liver diseases (CLD). Here, we examine "the gut homing theory" in patients with PSC with associated inflammatory bowel disease (PSC-IBD) and across multiple inflammatory liver diseases. APPROACH AND RESULTS: Expression of MAdCAM-1, CCL25, and E-Cadherin were assessed histologically and using RT-PCR on explanted liver tissue from patients with CLD undergoing OLT and in normal liver. Liver mononuclear cells were isolated from explanted tissue samples and the expression of gut homing integrins and cytokines on hepatic infiltrating gut-derived T cells was assessed using flow cytometry. Hepatic expression of MAdCAM-1, CCL25 and E-Cadherin was up-regulated in all CLDs compared with normal liver. There were no differences between disease groups. Frequencies of α4ß7, αEß7, CCR9, and GPR15 expressing hepatic T cells was increased in PSC-IBD, but also in CLD controls, compared with normal liver. ß7 expressing hepatic T cells displayed an increased inflammatory phenotype compared with ß7 negative cells, although this inflammatory cytokine profile was present in both the inflamed and normal liver. CONCLUSIONS: These findings refute the widely accepted "gut homing" hypothesis as the primary driver of PSC and indicate that aberrant hepatic recruitment of gut-derived T cells is not unique to PSC, but is a panetiological feature of CLD.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Cadherinas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Quimiocinas CC/metabolismo , Colangitis Esclerosante , Tracto Gastrointestinal , Hepatopatías , Hígado , Mucoproteínas/metabolismo , Moléculas de Adhesión Celular/aislamiento & purificación , Colangitis Esclerosante/inmunología , Colangitis Esclerosante/metabolismo , Colangitis Esclerosante/patología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Cadenas beta de Integrinas/metabolismo , Hígado/metabolismo , Hígado/patología , Hepatopatías/clasificación , Hepatopatías/metabolismo , Hepatopatías/patología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo
15.
Cardiovasc Diabetol ; 22(1): 84, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37046242

RESUMEN

AIM: Obesity is linked to cardiometabolic diseases, however non-obese individuals are also at risk for type 2 diabetes (T2D) and cardiovascular disease (CVD). White adipose tissue (WAT) is known to play a role in both T2D and CVD, but the contribution of WAT inflammatory status especially in non-obese patients with cardiometabolic diseases is less understood. Therefore, we aimed to find associations between WAT inflammatory status and cardiometabolic diseases in non-obese individuals. METHODS: In a population-based cohort containing non-obese healthy (n = 17), T2D (n = 16), CVD (n = 18), T2D + CVD (n = 19) individuals, seventeen different cytokines were measured in WAT and in circulation. In addition, 13-color flow cytometry profiling was employed to phenotype the immune cells. Human T cell line (Jurkat T cells) was stimulated by rCCL18, and conditioned media (CM) was added to the in vitro cultures of human adipocytes. Lipolysis was measured by glycerol release. Blocking antibodies against IFN-γ and TGF-ß were used in vitro to prove a role for these cytokines in CCL18-T-cell-adipocyte lipolysis regulation axis. RESULTS: In CVD, T2D and CVD + T2D groups, CCL18 and CD4+ T cells were upregulated significantly compared to healthy controls. WAT CCL18 secretion correlated with the amounts of WAT CD4+ T cells, which also highly expressed CCL18 receptors suggesting that WAT CD4+ T cells are responders to this chemokine. While direct addition of rCCL18 to mature adipocytes did not alter the adipocyte lipolysis, CM from CCL18-treated T cells increased glycerol release in in vitro cultures of adipocytes. IFN-γ and TGF-ß secretion was significantly induced in CM obtained from T cells treated with CCL18. Blocking these cytokines in CM, prevented CM-induced upregulation of adipocyte lipolysis. CONCLUSION: We suggest that in T2D and CVD, increased production of CCL18 recruits and activates CD4+ T cells to secrete IFN-γ and TGF-ß. This, in turn, promotes adipocyte lipolysis - a possible risk factor for cardiometabolic diseases.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Glicerol/metabolismo , Linfocitos T/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Citocinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/metabolismo , Quimiocinas CC/metabolismo
16.
Exp Dermatol ; 32(4): 379-391, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36398464

RESUMEN

Chemokines are a group of small proteins that induce chemoattraction and inflammation and contribute to the differentiation and homeostasis of various cell types. Here we explored the role of chemokines, extracellular matrix production, and myofibroblast differentiation in self-assembled skin equivalents (SASE), a three-dimensional (3D) skin-equivalent tissue model. We found that the expression of three chemokines, C-C motif chemokine ligand (CCL) 20, C-X-C motif chemokine ligand (CXCL) 5, and CXCL8, increased with differentiation to myofibroblasts. Addition of recombinant CCL20 to human skin fibroblast induced collagen Type I alpha 2 gene expression, but did not affect the expression of alpha smooth muscle actin expression. Conversely, siRNA gene knockdown of CCL20 effectively inhibited the expression of collagen Type I gene and protein. Furthermore, when the CCL20 gene in fibroblasts was knocked down in SASE, collagen Type I synthesis and stromal thickness were decreased. Taken together, these results have indicated the utility of SASE in understanding how cytokines such as CCL20 positively regulate extracellular matrix proteins such as collagen Type I production during myofibroblast differentiation in 3D tissues that mimic human skin.


Asunto(s)
Quimiocinas CC , Colágeno Tipo I , Humanos , Quimiocinas CC/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Ligandos , Piel/metabolismo , Fibroblastos/metabolismo , Miofibroblastos/metabolismo , Diferenciación Celular/fisiología , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Células Cultivadas , Actinas/metabolismo
17.
Cell Mol Life Sci ; 79(3): 155, 2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35218410

RESUMEN

Cellular senescence is closely related to tissue aging including bone. Bone homeostasis is maintained by the tight balance between bone-forming osteoblasts and bone-resorbing osteoclasts, but it undergoes deregulation with age, causing age-associated osteoporosis, a main cause of which is osteoblast dysfunction. Oxidative stress caused by the accumulation of reactive oxygen species (ROS) in bone tissues with aging can accelerate osteoblast senescence and dysfunction. However, the regulatory mechanism that controls the ROS-induced senescence of osteoblasts is poorly understood. Here, we identified Peptidyl arginine deiminase 2 (PADI2), a post-translational modifying enzyme, as a regulator of ROS-accelerated senescence of osteoblasts via RNA-sequencing and further functional validations. PADI2 downregulation by treatment with H2O2 or its siRNA promoted cellular senescence and suppressed osteoblast differentiation. CCL2, 5, and 7 known as the elements of the senescence-associated secretory phenotype (SASP) which is a secretome including proinflammatory cytokines and chemokines emitted by senescent cells and a representative feature of senescence, were upregulated by H2O2 treatment or Padi2 knockdown. Furthermore, blocking these SASP factors with neutralizing antibodies or siRNAs alleviated the senescence and dysfunction of osteoblasts induced by H2O2 treatment or Padi2 knockdown. The elevated production of these SASP factors was mediated by the activation of NFκB signaling pathway. The inhibition of NFκB using the pharmacological inhibitor or siRNA effectively relieved H2O2 treatment- or Padi2 knockdown-induced senescence and osteoblast dysfunction. Together, our study for the first time uncover the role of PADI2 in ROS-accelerated cellular senescence of osteoblasts and provide new mechanistic and therapeutic insights into excessive ROS-promoted cellular senescence and aging-related bone diseases.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Quimiocinas CC/metabolismo , Peróxido de Hidrógeno/farmacología , FN-kappa B/metabolismo , Arginina Deiminasa Proteína-Tipo 2/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL5/antagonistas & inhibidores , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CCL7/antagonistas & inhibidores , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Quimiocinas CC/antagonistas & inhibidores , Quimiocinas CC/genética , Daño del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Arginina Deiminasa Proteína-Tipo 2/antagonistas & inhibidores , Arginina Deiminasa Proteína-Tipo 2/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
18.
J Biol Chem ; 296: 100373, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33548230

RESUMEN

Mouse Ccr1l1 (Ccr1-like 1) encodes an orphan G-protein-coupled receptor (GPCR) with the highest homology to the inflammatory and highly promiscuous chemokine receptors Ccr1 and Ccr3 (70 and 50% amino acid identity, respectively). Ccr1l1 was first cloned in 1995, yet current knowledge of this putative chemokine receptor is limited to its gene organization and chromosomal localization. Here we report that Ccr1l1 is a Rodentia-specific gene selectively expressed in eosinophils. However, eosinophil phenotypes, development, and responsiveness to chemokines were all normal in naïve Ccr1l1 knockout mice. We demonstrate for the first time that recombinant Ccr1l1 is expressed on the plasma membrane of transfected cells and contains an extracellular N terminus and an intracellular C terminus, consistent with GPCR topology. Using receptor internalization, ß-arrestin recruitment, calcium flux, and chemotaxis assays, we excluded all 37 available mouse chemokines, including Ccr1 ligands, and two viral chemokines as Ccr1l1 ligands, and demonstrated that mouse Ccr1, but not Ccr1l1, exhibits constitutive signaling activity. However, sequence analysis and structural modeling revealed that Ccr1l1 is well equipped to act as a classical signaling GPCR, with N-terminal sulfotyrosines as the only signaling and chemokine-binding determinant absent in Ccr1l1. Hereof, we show that a sulfatable N-terminal Ccr1 Y18 residue is essential for chemotaxis and calcium responses induced by Ccl3 and Ccl9/10, but substituting the corresponding Ccr1l1 F19 residue with tyrosine failed to confer responsiveness to Ccr1 ligands. Although Ccr1l1 remains an extreme outlier in the chemokine receptor family, our study supports that it might respond to unidentified mouse chemokine ligands in eosinophil-driven immune responses.


Asunto(s)
Receptores CCR1/metabolismo , Animales , Membrana Celular/metabolismo , Quimiocinas/metabolismo , Quimiocinas CC/metabolismo , Quimiotaxis de Leucocito , Eosinófilos/metabolismo , Femenino , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores CCR1/fisiología , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Roedores/genética , Transducción de Señal , Relación Estructura-Actividad
19.
Kidney Int ; 102(4): 691-694, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36150760

RESUMEN

Kidney fibrosis is a hallmark of chronic kidney disease yet is poorly treated. Chang et al. determined that plasma and kidney levels of the vascular growth factor, angiopoietin-2, were elevated in patients with chronic kidney disease and mice with kidney disease. Angiopoietin-2 inhibited the renoprotective effects of angiopoietin-1 and promoted CC chemokine ligand 2-mediated kidney damage, endothelial cell apoptosis, vascular rarefaction, inflammation, fibrosis, and kidney dysfunction. Hence, therapeutically inhibiting angiopoietin-2 may represent a novel means of treating these chronic kidney disease-associated pathologies.


Asunto(s)
Angiopoyetina 2 , Insuficiencia Renal Crónica , Angiopoyetina 1 , Angiopoyetina 2/metabolismo , Animales , Quimiocinas CC/metabolismo , Fibrosis , Riñón/patología , Ligandos , Ratones , Insuficiencia Renal Crónica/patología
20.
Br J Cancer ; 127(6): 1026-1033, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35750747

RESUMEN

BACKGROUND: Macrophages are an important component of the tumour immune microenvironment (TME) and can promote tumour growth and metastasis. Macrophage-secreted chemokine-ligand-23 (CCL23) induces ovarian cancer cell migration via chemokine-receptor 1 (CCR1). However, the effect of CCL23 on other immune cells in the TME is unknown. METHODS: CCL23 levels were measured by ELISA. The expression of surface markers in exhaustion assays was quantified by flow cytometry. Signalling pathways were identified by phosphokinase array and validated by western blot. RESULTS: Ascites from patients with high-grade serous ovarian cancer (HGSC) contain high levels of CCL23. Similarly, significantly higher CCL23 levels were found in plasma from HGSC patients compared to healthy individuals. RNA-seq analysis of ovarian cancer tissues from TCGA showed that expression of CCL23 correlated with the presence of macrophages. In tissues with high levels of CCL23 and macrophage content, the fraction of CD8 + T cells expressing exhaustion markers CTLA-4 and PD-1 were significantly higher compared to low-level CCL23 tissues. In vitro, CCL23 induced upregulation of immune checkpoint proteins on CD8 + T cells, including CTLA-4, TIGIT, TIM-3 and LAG-3 via phosphorylation of GSK3ß in CD8 + T cells. CONCLUSIONS: Our data suggest that CCL23 produced by macrophages contributes to the immune-suppressive TME in ovarian cancer by inducing an exhausted T-cell phenotype.


Asunto(s)
Quimiocinas CC/metabolismo , Macrófagos/metabolismo , Neoplasias Ováricas , Microambiente Tumoral , Antígeno CTLA-4 , Carcinoma Epitelial de Ovario/metabolismo , Quimiocina CCL22/metabolismo , Femenino , Humanos , Ligandos , Neoplasias Ováricas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA