Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 16(7): e1008701, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32735617

RESUMEN

Gammaherpesviruses have evolved various strategies to take advantage of host cellular factors or signaling pathways to establish a lifelong latent infection. Like the human gammaherpesvirus Epstein-Barr virus, murine gammaherpesvirus 68 (MHV68) establishes and maintains latency in the memory B cells during infection of laboratory mice. We have previously shown that MHV68 can immortalize fetal liver-derived B cells that induce lymphomas when injected into immunodeficient mice. Here we identify interleukin 16 (IL16) as a most abundantly expressed cytokine in MHV68-immortalized B cells and show that MHV68 infection elevates IL16 expression. IL16 is not important for MHV68 lytic infection but plays a critical role in MHV68 reactivation from latency. IL16 deficiency increases MHV68 lytic gene expression in MHV68-immortalized B cells and enhances reactivation from splenic latency. Correlatively, IL16 deficiency increases the frequency of MHV68-infected plasma cells that can be attributed to enhanced MHV68 reactivation. Furthermore, similar to TPA-mediated lytic replication of Kaposi's sarcoma-associated herpesvirus, IL16 deficiency markedly induces Tyr705 STAT3 de-phosphorylation and elevates p21 expression, which can be counteracted by the tyrosine phosphatase inhibitor orthovanadate. Importantly, orthovanadate strongly blocks MHV68 lytic gene expression mediated by IL16 deficiency. These data demonstrate that virus-induced IL16 does not directly participate in MHV68 lytic replication, but rather inhibits virus reactivation to facilitate latent infection, in part through the STAT3-p21 axis.


Asunto(s)
Infecciones por Herpesviridae/metabolismo , Interleucina-16/metabolismo , Infecciones Tumorales por Virus/metabolismo , Activación Viral/fisiología , Latencia del Virus/fisiología , Animales , Linfocitos B/virología , Infecciones por Herpesviridae/inmunología , Interleucina-16/inmunología , Linfoma/virología , Ratones , Rhadinovirus/inmunología , Rhadinovirus/metabolismo
2.
Biol Pharm Bull ; 45(11): 1616-1626, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36328497

RESUMEN

The dysregulation of certain long non-coding RNAs (lncRNAs) has been considered to be involved in neuropsychiatric disorders such as depression, implying the vital role of these transcripts. We have previously identified many differentially expressed lncRNAs in chronic unpredictable mild stress (CUMS) induced mice. Among them, lncRNA Gm16638-201 was highly expressed in the hippocampus (HIP) of CUMS, but the specific role and the underlying mechanisms remain unclear. Here, we reported that lncRNA Gm16638-201 was highly expressed in the prefrontal cortex (PFC) of CUMS induced depressive mice. Bioinformatic analysis shows that Gm16638-201 is mainly located in the cytoplasm. Nine neurological-related genes (Elmo2, Satb1, Hnrnpul1, Sipa1l3, Mapt, Tada3, Sgip1, IL-16, and StarD5) were predicted to be regulated in cis or trans by Gm16638-201 and involved into the 14-3-3Ɛ neurotrophic signaling pathway. We further confirmed the down-regulation of 14-3-3Ɛ and the nine predicted target genes in the PFC of CUMS mice except for Sgip1 and IL-16. In addition, they were also down-regulated in the primary cortical cell cultures with overexpression of Gm16638-201 constructed using an adenoviral-medicated gene expression system. In conclusion, we found that overexpression of Gm16638-201 negatively regulated several target genes and inhibited the 14-3-3Ɛ pathway in the PFC of CUMS induced depressive mice. This promising result suggests that Gm16638-201 may be a potential novel therapeutic target for depression.


Asunto(s)
Antidepresivos , ARN Largo no Codificante , Ratones , Animales , Antidepresivos/uso terapéutico , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Depresión/tratamiento farmacológico , Estrés Psicológico/metabolismo , Interleucina-16/metabolismo , Modelos Animales de Enfermedad , Corteza Prefrontal/metabolismo , Hipocampo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción/metabolismo
3.
Cytokine ; 138: 155406, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33341000

RESUMEN

OBJECTIVES: To investigate the distribution of multiple cytokines in gastroschisis and reveal its association with clinical outcomes, including gastrointestinal disorders and fetal brain damage caused by chronic inflammation in gastroschisis. METHODS: We obtained amniotic fluid and arterial cord blood from 10 patients with gastroschisis, and evaluated the profile of 40 cytokines via multiplex immunoassay. The possible relationship of the cytokines with the time taken to attain full enteral nutrition and cord S100B, a surrogate marker of brain damage, was estimated. Associations among the relevant cytokines were also assessed. RESULTS: Although clinical characteristics in our cohort had no relevance, several cytokines in cord blood, especially IL-2, IL-8, CCL1, CCL7, CXCL1, CXCL2, and CXCL6, were clearly elevated in patients who took a longer time to attain full enteral nutrition, whereas only IL-16 in cord blood was significantly related to cord S100B and strongly correlation with cord S100B levels. Moreover, our data indicated that IL-16 was considerably less correlated with the other cytokines associated with adverse outcomes. CONCLUSIONS: We investigated the cytokine characteristics of both amniotic fluid and cord blood in gastroschisis, and found that certain cytokines could affect the adverse outcomes, including fetal brain damage. These findings provide important information that could further clarify the pathophysiology of gastroschisis and propose a novel clinical implication of gastroschisis that could be used to predict adverse outcomes, especially neurodevelopmental disorders.


Asunto(s)
Lesiones Encefálicas/embriología , Citocinas/metabolismo , Gastrosquisis/metabolismo , Adulto , Líquido Amniótico/metabolismo , Biomarcadores/metabolismo , Nutrición Enteral , Femenino , Sangre Fetal/metabolismo , Humanos , Recién Nacido , Inflamación , Interleucina-16/metabolismo , Edad Materna , Estudios Retrospectivos , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Adulto Joven
4.
BMC Cancer ; 21(1): 492, 2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-33941102

RESUMEN

BACKGROUND: Mycosis fungoides (MF) is a primary cutaneous T-cell lymphoma (CTCL) that transforms from mature, skin-homing T cells and progresses during the early stages in the skin. The role of the skin microenvironment in MF development is unclear, but recent findings in a variety of cancers have highlighted the role of stromal fibroblasts in promoting or inhibiting tumorigenesis. Stromal fibroblasts are an important part of the cutaneous tumor microenvironment (TME) in MF. Here we describe studies into the interaction of TME-fibroblasts and malignant T cells to gain insight into their role in CTCL. METHODS: Skin from normal (n = 3) and MF patients (n = 3) were analyzed for FAPα by immunohistochemistry. MyLa is a CTCL cell line that retains expression of biomarkers TWIST1 and TOX that are frequently detected in CTCL patients. MyLa cells were cultured in the presence or absence of normal or MF skin derived fibroblasts for 5 days, trypsinized to detached MyL a cells, and gene expression analyzed by RT-PCR for MF biomarkers (TWIST1 and TOX), Th1 markers (IFNG, TBX21), Th2 markers (GATA3, IL16), and proliferation marker (MKI67). Purified fibroblasts were assayed for VIM and ACTA2 gene expression. Cellular senescence assay was performed to assess senescence. RESULTS: MF skin fibroblast showed increased expression of FAP-α with increasing stage compared to normal. Normal fibroblasts co-cultured with MyLa cells suppressed expression of TWIST1 (p < 0.0006), and TOX (p < 0.03), GATA3 (p < 0.02) and IL16 (p < 0.03), and increased expression of IFNG (p < 0.03) and TBX21 (p < 0.03) in MyLa cells. In contrast, MyLa cells cultured with MF fibroblasts retained high expression of TWIST1, TOX and GATA3. MF fibroblasts co-culture with MyLa cells increased expression of IL16 (p < 0.01) and IL4 (p < 0.02), and suppressed IFNG and TBX21 in MyLa cells. Furthermore, expression of MKI67 in MyLa cells was suppressed by normal fibroblasts compared to MF fibroblasts. CONCLUSION: Skin fibroblasts represent important components of the TME in MF. In co-culture model, normal and MF fibroblasts have differential influence on T-cell phenotype in modulating expression of Th1 cytokine and CTCL biomarker genes to reveal distinct roles with implications in MF progression.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Linfoma Cutáneo de Células T/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/metabolismo , Microambiente Tumoral , Proteína 1 Relacionada con Twist/metabolismo , Actinas/genética , Actinas/metabolismo , Anciano , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Senescencia Celular , Técnicas de Cocultivo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-16/genética , Interleucina-16/metabolismo , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Linfoma Cutáneo de Células T/genética , Linfoma Cutáneo de Células T/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Micosis Fungoide/genética , Micosis Fungoide/metabolismo , Micosis Fungoide/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/citología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T/metabolismo , Proteína 1 Relacionada con Twist/genética , Vimentina/genética , Vimentina/metabolismo
5.
Gastroenterology ; 156(8): 2297-2312, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30836096

RESUMEN

BACKGROUND & AIMS: Interleukin 6 (IL6) and tumor necrosis factor contribute to the development of colitis-associated cancer (CAC). We investigated these signaling pathways and the involvement of G protein subunit alpha i1 (GNAI1), GNAI2, and GNAI3 in the development of CAC in mice and humans. METHODS: B6;129 wild-type (control) or mice with disruption of Gnai1, Gnai2, and/or Gnai3 or conditional disruption of Gnai2 in CD11c+ or epithelial cells were given dextran sulfate sodium (DSS) to induce colitis followed by azoxymethane (AOM) to induce carcinogenesis; some mice were given an antibody against IL6. Feces were collected from mice, and the compositions of microbiomes were analyzed by polymerase chain reactions. Dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) isolated from spleen and colon tissues were analyzed by flow cytometry. We performed immunoprecipitation and immunoblot analyses of colon tumor tissues, MDSCs, and mouse embryonic fibroblasts to study the expression levels of GNAI1, GNAI2, and GNAI3 and the interactions of GNAI1 and GNAI3 with proteins in the IL6 signaling pathway. We analyzed the expression of Gnai2 messenger RNA by CD11c+ cells in the colonic lamina propria by PrimeFlow, expression of IL6 in DCs by flow cytometry, and secretion of cytokines in sera and colon tissues by enzyme-linked immunosorbent assay. We obtained colon tumor and matched nontumor tissues from 83 patients with colorectal cancer having surgery in China and 35 patients with CAC in the United States. Mouse and human colon tissues were analyzed by histology, immunoblot, immunohistochemistry, and/or RNA-sequencing analyses. RESULTS: GNAI1 and GNAI3 (GNAI1;3) double-knockout (DKO) mice developed more severe colitis after administration of DSS and significantly more colonic tumors than control mice after administration of AOM plus DSS. Development of increased tumors in DKO mice was not associated with changes in fecal microbiomes but was associated with activation of nuclear factor (NF) κB and signal transducer and activator of transcription (STAT) 3; increased levels of GNAI2, nitric oxide synthase 2, and IL6; increased numbers of CD4+ DCs and MDSCs; and decreased numbers of CD8+ DCs. IL6 was mainly produced by CD4+/CD11b+, but not CD8+, DCs in DKO mice. Injection of DKO mice with a blocking antibody against IL6 reduced the expansion of MDSCs and the number of tumors that developed after CAC induction. Incubation of MDSCs or mouse embryonic fibroblasts with IL6 induced activation of either NF-κB by a JAK2-TRAF6-TAK1-CHUK/IKKB signaling pathway or STAT3 by JAK2. This activation resulted in expression of GNAI2, IL6 signal transducer (IL6ST, also called GP130) and nitric oxide synthase 2, and expansion of MDSCs; the expression levels of these proteins and expansion of MDSCs were further increased by the absence of GNAI1;3 in cells and mice. Conditional disruption of Gnai2 in CD11c+ cells of DKO mice prevented activation of NF-κB and STAT3 and changes in numbers of DCs and MDSCs. Colon tumor tissues from patients with CAC had reduced levels of GNAI1 and GNAI3 and increased levels of GNAI2 compared with normal tissues. Further analysis of a public human colorectal tumor DNA microarray database (GSE39582) showed that low Gani1 and Gnai3 messenger RNA expression and high Gnai2 messenger RNA expression were significantly associated with decreased relapse-free survival. CONCLUSIONS: GNAI1;3 suppresses DSS-plus-AOM-induced colon tumor development in mice, whereas expression of GNAI2 in CD11c+ cells and IL6 in CD4+/CD11b+ DCs appears to promote these effects. Strategies to induce GNAI1;3, or block GNAI2 and IL6, might be developed for the prevention or therapy of CAC in patients.


Asunto(s)
Transformación Celular Neoplásica/genética , Colitis/patología , Neoplasias del Colon/patología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Animales , Biopsia con Aguja , Carcinogénesis , Colitis/genética , Neoplasias del Colon/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica , Interleucina-16/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Valores de Referencia , Sensibilidad y Especificidad , Transducción de Señal/genética
6.
Am J Pathol ; 189(1): 132-146, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30553437

RESUMEN

Cartilage oligomeric matrix protein (COMP) is a large, multifunctional extracellular protein that, when mutated, is retained in the rough endoplasmic reticulum (ER). This retention elicits ER stress, inflammation, and oxidative stress, resulting in dysfunction and death of growth plate chondrocytes. While identifying the cellular pathologic mechanisms underlying the murine mutant (MT)-COMP model of pseudoachondroplasia, increased midline-1 (MID1) expression and mammalian target of rapamycin complex 1 (mTORC1) signaling was found. This novel role for MID1/mTORC1 signaling was investigated since treatments shown to repress the pathology also reduced Mid1/mTORC1. Although ER stress-inducing drugs or tumor necrosis factor α (TNFα) in rat chondrosarcoma cells increased Mid1, oxidative stress did not, establishing that ER stress- or TNFα-driven inflammation alone is sufficient to elevate MID1 expression. Since MID1 ubiquitinates protein phosphatase 2A (PP2A), a negative regulator of mTORC1, PP2A was evaluated in MT-COMP growth plate chondrocytes. PP2A was decreased, indicating de-repression of mTORC1 signaling. Rapamycin treatment in MT-COMP mice reduced mTORC1 signaling and intracellular retention of COMP, and increased proliferation, but did not change inflammatory markers IL-16 and eosinophil peroxidase. Lastly, mRNA from tuberous sclerosis-1/2-null mice brain tissue exhibiting ER stress had increased Mid1 expression, confirming the relationship between ER stress and MID1/mTORC1 signaling. These findings suggest a mechanistic link between ER stress and MID1/mTORC1 signaling that has implications extending to other conditions involving ER stress.


Asunto(s)
Acondroplasia , Proteína de la Matriz Oligomérica del Cartílago , Sistemas de Liberación de Medicamentos , Diana Mecanicista del Complejo 1 de la Rapamicina , Acondroplasia/tratamiento farmacológico , Acondroplasia/genética , Acondroplasia/patología , Animales , Biomarcadores/metabolismo , Proteína de la Matriz Oligomérica del Cartílago/genética , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Línea Celular Tumoral , Condrocitos/metabolismo , Condrocitos/patología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Retículo Endoplásmico Rugoso/genética , Retículo Endoplásmico Rugoso/metabolismo , Retículo Endoplásmico Rugoso/patología , Peroxidasa del Eosinófilo/genética , Peroxidasa del Eosinófilo/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-16/genética , Interleucina-16/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas/genética , Proteínas/metabolismo , Ratas , Transducción de Señal/genética , Sirolimus/farmacología , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas
7.
Rheumatology (Oxford) ; 59(3): 519-523, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31377804

RESUMEN

OBJECTIVES: SSc is an autoimmune disease with chronic and persistent inflammation in its pathogenesis. To examine the expression pattern of IL-16 in SSc lesions, the serum concentration of IL-16 in SSc patients and the relationship between serum IL-16 levels and the clinical symptoms of SSc were investigated. METHODS: Using immunohistochemical analysis, we examined the quantity and localization of IL-16 in affected skin obtained from SSc patients. We also measured serum levels of IL-16 in SSc patients using an ELISA. We then validated the correlation between serum IL-16 levels and clinical symptoms in patients with SSc. RESULTS: In the skin, IL-16 was expressed on the lymphocytes around the capillaries. Furthermore, the proportion of IL-16-positive cells was statistically higher in patients with dcSSc than in those with lcSSc patients (43.9 vs 29.1%, P < 0.05). The serum IL-16 levels in SSc patients were statistically significant elevated compared with healthy controls (297.0 vs 194.9 pg/ml, P < 0.05). Increased serum IL-16 levels in SSc patients were correlated with the proportion classified as dcSSc, skin score and the presence of cutaneous symptoms of erythema and pigmentation. CONCLUSION: The regional up-regulation of IL-16 in the skin is not only associated with skin sclerosis, but also with systemic IL-16 activation. IL-16 may play a role in the pathogenesis of SSc. Moreover, serum IL-16 levels may be useful as a biomarker for determining the severity of the skin sclerosis. Inhibiting IL-16 activation may be effective in treating SSc.


Asunto(s)
Interleucina-16/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/metabolismo , Adulto , Anciano , Biomarcadores/metabolismo , Capilares/metabolismo , Femenino , Humanos , Interleucina-16/sangre , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Esclerodermia Sistémica/sangre , Esclerodermia Sistémica/diagnóstico , Índice de Severidad de la Enfermedad
8.
Scand J Immunol ; 92(6): e12926, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32862475

RESUMEN

In the effort of developing new immunotherapies, the sentinel node (SN) has proven a promising source from which to harness an effective antitumour T cell response. However, tumour immune escape, a process in which regulatory T cells (Tregs) play a central role, remains a major limiting factor. Therefore, there is a clear need to increase the knowledge of Treg function and signalling in sentinel nodes. Here, we set out to explore whether the proteome in SN-resident T cells is altered by the tumour and to identify key proteins in SN T cell signalling, focusing on Tregs. Five patients with muscle-invasive urothelial bladder cancer were prospectively included. Mass spectrometry was performed on two patients, with validation and functional studies being performed on three additional patients and four healthy donors. At cystectomy, SN, non-SN lymph nodes and peripheral blood samples were collected from the patients and T cell subsets isolated through flow cytometry before downstream experiments. Proteomic analysis indicated that growth and immune signalling pathways are upregulated in SN-resident Tregs. Furthermore, centrality analysis identified the cytokine IL-16 to be central in the SN-Treg signalling network. We show that tumour-released factors, through activating caspase-3, increase Treg IL-16 processing into bioactive forms, reinforcing Treg suppressive capacity. In conclusion, we provide evidence that Tregs exposed to secreted factors from bladder tumours show increased immune and growth signalling and altered IL-16 processing which translates to enhanced Treg suppressive function, indicating altered IL-16 signalling as a novel tumour immune escape mechanism.


Asunto(s)
Interleucina-16/metabolismo , Neoplasias de los Músculos/inmunología , Ganglio Linfático Centinela/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Urotelio/patología , Anciano , Anciano de 80 o más Años , Femenino , Citometría de Flujo , Humanos , Masculino , Neoplasias de los Músculos/secundario , Estadificación de Neoplasias , Proteómica , Transducción de Señal , Escape del Tumor , Neoplasias de la Vejiga Urinaria/patología
9.
Int J Mol Sci ; 21(8)2020 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-32326301

RESUMEN

Because of lipopolysaccharide (LPS)-mediated effects on osteoclast differentiation and bone loss, periprosthetic joint infection (PJI) caused by Gram-negative bacteria increases the risk of aseptic loosening after reimplantation. Synovial fluid interleukin-16 (IL-16) expression was higher in patients with PJI than in patients without joint infection. Thus, we explored the effects of IL-16 on bone. We investigated whether IL-16 modulates osteoclast or osteoblast differentiation in vitro. An LPS-induced bone loss mice model was used to explore the possible advantages of IL-16 inhibition for the prevention of bone loss. IL-16 directly activated p38 and c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) signaling and increased osteoclast activation markers, including tartrate-resistant acid phosphatase (TRAP), cathepsin K, and nuclear factor of activated T cells 1 (NFATc1). IL-16 directly caused monocytes to differentiate into TRAP-positive osteoclast-like cells through NFATc1 activation dependent on JNK/MAPK signaling. Moreover, IL-16 did not alter alkaline phosphatase activity or calcium deposition during osteoblastic differentiation. Finally, IL-16 inhibition prevented LPS-induced trabecular bone loss and osteoclast activation in vivo. IL-16 directly increased osteoclast activation through the JNK/NFATc1 pathway. IL-16 inhibition could represent a new strategy for treating infection-associated bone loss.


Asunto(s)
Artritis Infecciosa/metabolismo , Resorción Ósea/metabolismo , Interleucina-16/metabolismo , Sistema de Señalización de MAP Quinasas , Osteoclastos/metabolismo , Infecciones Relacionadas con Prótesis/metabolismo , Líquido Sinovial/metabolismo , Animales , Artritis Infecciosa/etiología , Biomarcadores , Catepsina K/genética , Catepsina K/metabolismo , Expresión Génica , Inmunohistoquímica , Interleucina-16/antagonistas & inhibidores , Lipopolisacáridos/inmunología , Ratones , Modelos Biológicos , Infecciones Relacionadas con Prótesis/microbiología , Células RAW 264.7
10.
Immunology ; 157(3): 257-267, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31120548

RESUMEN

Asthma is a chronic inflammatory disease that involves a variety of cytokines and cells. Interleukin-16 (IL-16) is highly expressed during allergic airway inflammation and is involved in its development. However, its specific mechanism of action remains unclear. In the present study, we used an animal model of ovalbumin (OVA)-induced allergic asthma with mice harboring an IL-16 gene deletion to investigate the role of this cytokine in asthma, in addition to its underlying mechanism. Increased IL-16 expression was observed during OVA-induced asthma in C57BL/6J mice. However, when OVA was used to induce asthma in IL-16-/- mice, a diminished inflammatory reaction, decreased bronchoalveolar lavage fluid (BALF) eosinophil numbers, and the suppression of OVA-specific IgE levels in the serum and BALF were observed. The results also demonstrated decreased levels of T helper type 2 (Th2) and Th17 cytokines upon OVA-induced asthma in IL-16-/- mice. Hence, we confirmed that IL-16 enhances the lung allergic inflammatory response and suggest a mechanism possibly associated with the up-regulation of IgE and the promotion of Th2 and Th17 cytokine production. This work explored the mechanism underlying the regulation of IL-16 in asthma and provides a new target for the clinical treatment of asthma.


Asunto(s)
Asma/metabolismo , Hiperreactividad Bronquial/metabolismo , Interleucina-16/metabolismo , Pulmón/metabolismo , Ovalbúmina , Células Th17/metabolismo , Células Th2/metabolismo , Animales , Asma/inmunología , Asma/fisiopatología , Asma/prevención & control , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Hiperreactividad Bronquial/prevención & control , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Broncoconstricción , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Interleucina-16/deficiencia , Interleucina-16/genética , Pulmón/inmunología , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Células Th17/inmunología , Células Th2/inmunología
11.
Cancer Sci ; 110(10): 3068-3078, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31432577

RESUMEN

The programmed cell death 1/programmed cell death 1 ligand 1 pathway was successfully targeted in cancer immunotherapy. Elevated interleukin-17 (IL-17), which is known in autoimmune diseases, has recently been recognized in cancer patients. We investigated the role of IL-17 in the regulation of expression of programmed cell death 1 ligand 1 in ovarian cancer by evaluating changes in the number of IL-17-producing cluster of differentiation 4 helper T cells (Th17) and γδT cells (γδT17) in PBMC of 52 gynecological cancer patients (including 30 ovarian cancer patients) and 18 healthy controls. The occupancy ratio of Th17 and γδT17 was higher in ovarian cancer and endometrial cancer patients than in controls, determined by multi-color flow cytometry (Th17: P < 0.0001 and P = 0.0002, respectively; γδT17: P = 0.0020 and P = 0.0084, respectively). IL-17 mRNA level was elevated in PBMC of ovarian cancer patients (P = 0.0029), as measured by RT-PCR. The neutrophil-to-lymphocyte ratio, which is a prognostic biomarker of ovarian cancer, correlated with Th17 occupancy ratio in patients (P = 0.0068). We found that programmed cell death 1 ligand 1 expression and its associated factors (IL-6 and phospho-signal transducer and activator of transcription 3) were induced by IL-17 in an ovarian cancer cell line. These results suggest that increased Th17 counts and IL-17 level, which correlated with high neutrophil-to-lymphocyte ratio and programmed cell death 1 ligand 1 expression, are potential biomarkers for poor prognosis in ovarian cancer and likely indications for application of programmed cell death 1 ligand 1 pathway inhibitors.


Asunto(s)
Antígeno B7-H1/genética , Neoplasias Endometriales/genética , Interleucina-16/metabolismo , Interleucina-17/genética , Neoplasias Ováricas/genética , Factor de Transcripción STAT3/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Línea Celular Tumoral , Neoplasias Endometriales/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/metabolismo , Neoplasias Ováricas/inmunología , Fosforilación , Pronóstico , Células Th17/metabolismo , Regulación hacia Arriba
12.
Fish Shellfish Immunol ; 88: 284-292, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30849500

RESUMEN

SAHH is an enzyme, playing a significant role in the catalyzation of the S-adenosyl homocysteine (SAH) into homocysteine (Hcy) and adenosine (Ado). However, little is known information of the enzyme in crustaceans. In the present study, SAHH cDNA was cloned from Litopenaeus vannamei (LvSAHH). The full length of the LvSAHH was found, containing a 5' UTR of 119 bp, an ORF of 1236 bp and a 3' UTR of 549 bp. The LvSAHH gene encoded a polypeptide of 411 amino acids with an estimated molecular mass of 45.55 kD and a predicted isoelectronic point (pI) of 5.63. Comparison of the deduced amino acid sequence showed that LvSAHH has high identity (70 %-82%) with other known species. qRT-PCR analysis revealed that LvSAHH mRNA was broadly expressed in all of the examined tissues, while the highest expression level was observed in muscle, followed by the expression in stomach, gill, pleopod, hepatopancreas, heart, eye and intestine. Subcellular localization analysis revealed that LvSAHH was predominantly localized in the cytoplasm and nucleus. LvSAHH mRNA expression levels in hepatopancreas and gill were significantly up-regulated from 6 to 48 h after V. alginolyticus injection and reached the highest level (15-fold and 8-fold, p < 0.01) at 24 h, respectively. Additionally, the Toll-like receptors (TLR) and interleukins-16 (IL-16) were detected in hepatopancreas and gill of LvSAHH-knockdown SAHH. LvRack1, LvToll1, LvToll2, LvToll3 and LvIL-16 transcripts were decreased significantly in LvSAHH-knockdown shrimp at 24 h post V. alginolyticus stimulation in hepatopancreas and gill. But LvToll3 was no significant difference in gill. In summary, these results indicated that LvSAHH may play a regulatory role in the invertebrate innate immune defense by regulating TLR and IL-16 expression.


Asunto(s)
Adenosilhomocisteinasa/metabolismo , Penaeidae/inmunología , Vibrio alginolyticus , Adenosilhomocisteinasa/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Expresión Génica , Técnicas de Silenciamiento del Gen , Inmunidad Innata/genética , Interleucina-16/metabolismo , Penaeidae/enzimología , Penaeidae/microbiología , ARN Mensajero/metabolismo , Receptores Toll-Like/metabolismo
13.
J Immunol ; 198(4): 1395-1402, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167650

RESUMEN

Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.


Asunto(s)
Alarminas/fisiología , Estrés Fisiológico , Animales , Proteína HMGB1/metabolismo , Humanos , Inflamación , Interleucina-16/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-33/metabolismo , Ratones , Procesamiento Proteico-Postraduccional , Estrés Fisiológico/inmunología , Estrés Fisiológico/fisiología
14.
Chembiochem ; 18(8): 721-725, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28186695

RESUMEN

The farnesoid X receptor (FXR) is an important target for drug discovery. Small molecules induce a conformational change in FXR that modulates its binding to co-regulators, thus resulting in distinct FXR functional profiles. However, the mechanisms for selectively recruiting co-regulators by FXR remain elusive, partly because of the lack of FXR-selective modulators. We report the identification of two natural terpenoids, tschimgine and feroline, as novel FXR modulators. Remarkably, their crystal structures uncovered a secondary binding pocket important for ligand binding. Further, tschimgine or feroline induced dynamic conformational changes in the activation function 2 (AF-2) surface, thus leading to differential co-regulator recruiting profiles, modulated by both hydrophobic and selective hydrogen-bond interactions unique to specific co-regulators. Our findings thus provide a novel structure template for optimization for FXR-selective modulators of clinical value.


Asunto(s)
Compuestos Bicíclicos con Puentes/farmacología , Ciclodecanos/farmacología , Hidroxibenzoatos/farmacología , Parabenos/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Animales , Sitios de Unión , Haplorrinos , Células Hep G2 , Humanos , Interleucina-16/metabolismo , Ligandos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Mutación Puntual , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/genética , Factor de Necrosis Tumoral alfa/metabolismo
15.
Ann Allergy Asthma Immunol ; 118(2): 191-196, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27986411

RESUMEN

BACKGROUND: Interleukin (IL) 16 and thymus and activation-regulated cytokine (TARC) are chemoattractant cytokines for eosinophils and TH2 cells. Differential levels of these components in aspirin-exacerbated respiratory disease (AERD) and allergic rhinitis with asthma (ARwA) may be related to a different inflammatory response in both asthma phenotypes. OBJECTIVE: To assess the nasal lavage immunoreactivity of IL-16 and TARC cytokines. METHODS: We used multienzyme-linked immunosorbent assays to detect IL-5, IL-13, IL-16, IL-33, I-309/CCL1, TARC/CCL17, monocyte-derived chemokine/CCL22, periostin, and eosinophil cationic protein levels in nasal lavages from patients with AERD and patients with ARwA. RESULTS: The IL-13, IL-16, TARC, and periostin levels were significantly higher in patients with AERD compared with those of patients with ARwA. Correlation analysis of mediator levels in AERD revealed a possible role of IL-16 and TARC in eosinophil recruitment and activation. CONCLUSION: IL-16, TARC, and periostin distinguish between patients with AERD and those with ARwA. These mediators, taken together rather than individually, may comprise good specific nasal markers in patients with AERD. The effects of IL-16 and TARC on TH1, TH2, and T-regulatory cell functions in AERD cannot be disregarded.


Asunto(s)
Aspirina/efectos adversos , Quimiocina CCL17/metabolismo , Hipersensibilidad a las Drogas/etiología , Hipersensibilidad a las Drogas/metabolismo , Interleucina-16/metabolismo , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/metabolismo , Adulto , Biomarcadores , Hipersensibilidad a las Drogas/diagnóstico , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Masculino , Persona de Mediana Edad , Líquido del Lavado Nasal/inmunología , Fenotipo , Pruebas de Función Respiratoria , Hipersensibilidad Respiratoria/diagnóstico , Pruebas Cutáneas , Células Th2/inmunología , Células Th2/metabolismo , Adulto Joven
16.
J Pathol ; 239(3): 262-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27037906

RESUMEN

Multiple myeloma (MM) is a chronic progressive malignancy of plasma cells. Although treatment with the novel proteasome inhibitor, bortezomib, significantly improves patient survival, some patients fail to respond due to the development of de novo resistance. We have previously shown that cytotoxic drugs can induce pro-tumorigenic host-mediated effects which contribute to tumour re-growth and metastasis, and thus limit anti-tumour efficacy. However, such effects and their impact on tumour cell aggressiveness have not been investigated using cytostatic agents such as bortezomib. Here we show that plasma from bortezomib-treated mice significantly increases migration, viability and proliferation of MM cells in vitro, compared to plasma from vehicle treated mice. In vivo, bortezomib induces the mobilization of pro-angiogenic bone marrow cells. Furthermore, mice treated with bortezomib and subsequently were used as recipients for an injection of MM cells succumb to MM earlier than mice treated with the vehicle. We show that bortezomib promotes pro-inflammatory macrophages which account for MM cell aggressiveness, an effect which is partially mediated by interleukin-16. Accordingly, co-inoculation of MM cells with pro-inflammatory macrophages from bortezomib-treated mice accelerates MM disease progression. Taken together, our results suggest that, in addition to the known effective anti-tumour activity of bortezomib, host-driven pro-tumorigenic effects generated in response to treatment can promote MM aggressiveness, and thus may contribute to the overall limited efficacy. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma/uso terapéutico , Inductores de la Angiogénesis , Animales , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Bortezomib/efectos adversos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-16/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mieloma Múltiple/patología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/patología , Inhibidores de Proteasoma/efectos adversos
17.
J Neuroinflammation ; 12: 78, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25896927

RESUMEN

In an important article published in Nature Medicine, Liu and colleagues described a novel CD4(+) FoxA1(+) regulatory T (Treg) cell population as distinct regulators of relapsing-remitting multiple sclerosis (RRMS) and experimental autoimmune encephalomyelitis (EAE). CD4(+) FoxA1(+) Treg cells appear as key regulators of responsiveness to therapy with interferon beta (IFN-ß) in RRMS patients. Data indicate that CD4(+)FoxA1(+) FOXP3(-) Treg cells develop within the central nervous system (CNS), and a potential of cerebellar granule neurons (CGN) in generation of CD4(+)FoxA1(+)PD-L1(hi)FOXP3(-) Treg cells from encephalitogenic CD4(+) T cells. A CD4 co-receptor specific ligand, IL-16, governs trafficking and biological properties of CD4(+) T cells irrespective of their activation state. Functions of IL-16, relevant to Treg cells, include expansion of CD4(+)CD25(+) T cells in long-term cultures with IL-2, de novo induction of FOXP-3 and migration of FOXP-3(+) T cells. IL-16 is highly conserved across species including human and mouse. CGN and neurons in hippocampus contain neuronal-IL-16 (NIL-16), splice variant of immune IL-16, and express CD4 molecule. In a CD4-dependent manner, IL-16 supports cultured CGN survival. Concomitant studies of RRMS lesions and corresponding MOG35-55-induced relapsing EAE in (B6 × JL)F1 (H-2(b/s)) mice discovered similar roles of IL-16 in regulation of relapsing disease. In RRMS and EAE relapse, peak levels of IL-16 and active caspase-3 correlated with CD4(+) T cell infiltration and levels of T-bet, Stat-1(Tyr(701)), and phosphorylated neurofilaments of axonal cytoskeleton [NF (M + H) P], suggesting a role of locally produced IL-16 in regulation of CD4(+) Th1 inflammation and axonal damage, respectively. IL-16 was abundantly present in CD4(+) T cells, followed by CD20(+) B, CD8(+) T, CD83(+) dendritic cells, and Mac-1(+) microglia. Apart from lesions, bioactive IL-16 was located in normal-appearing white matter (NAWM) and normal-appearing grey matter (NAGM) in RRMS brain and spinal cord. A cytokine IL-16 emerges as an important regulator of relapsing MS and EAE. Better understanding of immune cell-neuron interactions mediated by IL-16 will foster development of more specific CD4(+) T cell subset-targeted therapies to prevent or ameliorate progression of neuroinflammation and axonal and neuronal damage. Translational studies necessitate corresponding EAE models.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Interleucina-16/metabolismo , Esclerosis Múltiple Recurrente-Remitente/patología , Animales , Humanos , Ratones , Esclerosis Múltiple Recurrente-Remitente/inducido químicamente , Esclerosis Múltiple Recurrente-Remitente/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad
18.
Biol Reprod ; 92(3): 82, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25673564

RESUMEN

Considering that interleukin 10 (IL10), transforming growth factor beta1 (TGFB1), and interferon gamma (IFNG) are involved in the susceptibility of BeWo trophoblast cells to Toxoplasma gondii infection, the aim of the present study was to investigate the effector mechanisms triggered by these cytokines in the control of T. gondii in BeWo cells. For this purpose, infected/uninfected BeWo cells were treated with IL10, TGFB1 (50 ng/ml), and IFNG (20 or 100 ng/ml) in order to verify the phosphorylation of signal transducers and activators of transcription 1 (STAT1), STAT3, and Smad2, parasite intracellular proliferation, as well as the Th1/Th2/IL17A cytokine production. The treatment of BeWo cells with IL10 and TGFB1 favored T. gondii proliferation, and these findings were associated with STAT3 and Smad2 phosphorylation, respectively (P < 0.05). Also, these cytokine treatments were able to down-modulate TNF alpha (TNFA) and IL6 production (P < 0.05). Low concentration of IFNG was unable to control T. gondii infection but was able to trigger STAT1 phosphorylation and up-regulate IL6 and IL17A production; whereas a high concentration of IFNG was unable to activate STAT1 but down-modulated IL6 and TNFA and increased T. gondii proliferation (P < 0.05). IL10, TGFB1, and IFNG regulate a differential T. gondii proliferation in BeWo cells because they distinctly trigger intracellular signaling pathways and cytokine production, especially IL6 and TNFA. Our data open new windows to understand the mechanisms triggered by IL10, TGFB1, and IFNG at the maternal-fetal interface in the presence of T. gondii, contributing to recognizing the importance of these effector mechanisms involved in the vertical transmission of this parasite.


Asunto(s)
Citocinas/metabolismo , Interferón gamma/farmacología , Interleucina-10/farmacología , Transducción de Señal/efectos de los fármacos , Toxoplasmosis/prevención & control , Factor de Crecimiento Transformador beta1/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/parasitología , Línea Celular Tumoral , Coriocarcinoma/patología , Susceptibilidad a Enfermedades , Femenino , Humanos , Técnicas In Vitro , Interleucina-16/metabolismo , Fosforilación , Embarazo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Proteína Smad2/metabolismo , Toxoplasma/aislamiento & purificación , Toxoplasmosis/metabolismo , Toxoplasmosis/patología , Trofoblastos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Neoplasias Uterinas/patología
19.
Clin Endocrinol (Oxf) ; 83(5): 726-32, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25940130

RESUMEN

OBJECTIVES: Cytokines have an important role in orchestrating the pathophysiology in autoimmune thyroid disease. The aim of the current study was to analyse the expression of interleukin (IL)-14 and IL-16 in the thyroid tissue of patients with Graves' disease (GD), Hashimoto's thyroiditis (HT) or multinodular goitre (MNG) and in that of normal individuals, in patients' intrathyroidal CD4(+) and CD8(+) T cells, and in patient and normal cultured thyroid follicular cells. METHODS: The expression of IL-14 and IL-16 mRNA and protein was investigated using reverse transcription-polymerase chain reaction (RT-PCR) amplification, and Western blotting and ELISAs, respectively. RESULTS: IL-14 mRNA expression was detected in thyroid tissue from 8/9 GD, 3/4 HT and 3/13 MNG patients and 1/6 normal individuals, and IL-16 mRNA expression in thyroid tissue from 9/9 GD, 4/4 HT and 9/13 MNG patients and 4/6 normal individuals. IL-14 mRNA expression was detected in intrathyroidal CD4(+) and CD8(+) T cells from 2/2 GD and 2/2 HT patients, while IL-16 mRNA was detected in samples from 1/2 HT patients but not in those from either patient with GD. IL-14 and IL-16 mRNA expression was found in thyroid follicular cells derived from 2/2 patient with GD and 1/1 normal individual. IL-14 protein was detected in thyroid tissue from 6/6 GD, 1/1 HT and 0/6 MNG patients and 0/6 normal individuals, and IL-16 protein in thyroid tissue from 6/6 GD, 1/1 HT and 1/6 MNG patients and 0/6 normal individuals. Expression of IL-14 protein was stimulated in thyroid follicular cells derived from two patients with GD and one normal individual by peripheral blood mononuclear cell (PBMC)-conditioned medium. Treatment of thyrocytes from two patients with GD and one normal individual with PBMC-conditioned medium and tumour necrosis factor (TNF)-α stimulated IL-16 protein expression. In normal thyrocytes, IL-16 protein synthesis was induced also by IL-1ß, IL-17A, IL-4 and transforming growth factor (TGF)-ß. CONCLUSIONS: The data provide evidence that the intrathyroidal production of IL-14 and IL-16 is associated with the pathogenesis of autoimmune thyroid disease. Thyroid follicular cells display the ability to express IL-14 and IL-16 mRNA and can be stimulated to express IL-16 protein, by a panel of cytokines, and IL-14 protein, by as yet unidentified factors.


Asunto(s)
Enfermedad de Graves/metabolismo , Enfermedad de Hashimoto/metabolismo , Interleucina-16/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Estudios de Casos y Controles , Enfermedad de Graves/inmunología , Enfermedad de Hashimoto/inmunología , Humanos , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Tirotropina
20.
J Immunol ; 191(3): 1413-27, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23817423

RESUMEN

Inflammatory bowel disease (IBD) has long been a worldwide health care problem with a persistently increasing incidence. Although its clinical features have been well described, its etiology and pathogenesis remain unclear. IL-16 is a chemoattractant cytokine with various effects on cellular activities and diseases. However, the involvement of IL-16 in IBD remains poorly understood. In this study, to our knowledge we report for the first time the mechanism by which IL-16 induces intestinal inflammation by upregulating the expression of oligopeptide transporter member 1 (PepT1) in a Tetraodon nigroviridis fish model. The dextran sodium sulfate-induced colitis model in this species revealed that IL-16 levels significantly increase accompanied by elevations in PepT1 in the colon. Moreover, the signs of colitis were dramatically attenuated by IL-16 depletion using anti-IL-16 Abs. In vivo IL-16 administration induced remarkable intestinal inflammation with typical ulcerative colitis-like features, including histologic damage, inflammatory cell infiltration, increased myeloperoxidase activity, and proinflammatory cytokines expression, which corresponded with significant PepT1 upregulation in the colon. The IL-16-induced PepT1 expression and its upregulated fMLF transport were also demonstrated in vitro. To our knowledge, our study provides the first evidence of the connection between IL-16 and PepT1, which provides new insights into the molecular mechanism underlying IBD development. Additionally, this study suggests that fish species are an attractive model for studying IBD. By providing a better understanding of IL-16 biology from fish to mammals, this study should aid the development of IL-16-based therapies for IBD.


Asunto(s)
Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucina-16/metabolismo , Intestinos/inmunología , Simportadores/metabolismo , Animales , Anticuerpos/inmunología , Línea Celular Tumoral , Sulfato de Dextran , Modelos Animales de Enfermedad , Células HeLa , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Interleucina-16/inmunología , Interleucina-16/farmacología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transportador de Péptidos 1 , Interferencia de ARN , ARN Interferente Pequeño , Simportadores/genética , Tetraodontiformes/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA