Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nephron ; 148(6): 437-442, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38281481

RESUMEN

BACKGROUND: Chemokines orchestrate immune cells activation and infiltration during acute kidney injury (AKI). OBJECTIVES: We aim to test whether deletion of C-C chemokine ligand 7 (CCL7), a small chemokine related to CCL2 (MCP-1), may modulate AKI development and progression toward kidney fibrosis. METHOD: Expression of CCL7 was quantified in murine cortical tubular (MCT) cells exposed to myoglobin or lipopolysaccharide or submitted to metabolic reprogramming. Kidney function (BUN, glomerular filtration rate), expression of CCL7 receptors, and kidney infiltration by inflammatory cells (F4/80+ macrophages, MPO+ neutrophils, and B220+ B-cells) were assessed in wt and Ccl7-/- mice submitted to 3 different models of AKI or kidney fibrosis (uni/bilateral ischemia/reperfusion injury (u/bIRI) and rhabdomyolysis). RESULTS: Toxin exposure of MCT cells, as well as metabolic reprogramming recapitulating AKI changes, led to a dramatic up-regulation of CCL7. In vivo, kidney expression of Ccl7 and Ccl2 significantly increased after AKI and remained increased beyond the acute phase (30 days after uIRI). The expression of the CCL7 receptors was heterogeneous and varied with time. Kidney function, expression of CCL7 receptors and Ccl2, and the number of inflammatory cells within kidneys were similar in wt and Ccl7-/- mice at baseline and at day 2 after AKI. Thirty days after uIRI, kidney fibrosis was similar in both mouse strains. CONCLUSIONS: Despite strong induction of CCL7 after AKI, CCL7 deficiency does not prevent AKI and the transition toward kidney fibrosis and should probably not be further explored as a potential target to prevent or treat AKI.


Asunto(s)
Lesión Renal Aguda , Quimiocina CCL7 , Ratones Noqueados , Animales , Lesión Renal Aguda/metabolismo , Quimiocina CCL7/metabolismo , Quimiocina CCL7/genética , Ratones , Ratones Endogámicos C57BL , Biomarcadores/metabolismo , Daño por Reperfusión , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Masculino , Fibrosis , Riñón/patología , Riñón/metabolismo , Rabdomiólisis , Modelos Animales de Enfermedad
2.
Infect Immun ; 91(4): e0001423, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36880752

RESUMEN

Staphylococcus aureus is the principal causative agent of osteomyelitis, a serious bacterial infection of bone that is associated with progressive inflammatory damage. Bone-forming osteoblasts have increasingly been recognized to play an important role in the initiation and progression of detrimental inflammation at sites of infection and have been demonstrated to release an array of inflammatory mediators and factors that promote osteoclastogenesis and leukocyte recruitment following bacterial challenge. In the present study, we describe elevated bone tissue levels of the potent neutrophil-attracting chemokines CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7 in a murine model of posttraumatic staphylococcal osteomyelitis. RNA sequencing (RNA-Seq) gene ontology analysis of isolated primary murine osteoblasts showed enrichment in differentially expressed genes involved in cell migration and chemokine receptor binding and chemokine activity following S. aureus infection, and a rapid increase in the expression of mRNA encoding CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7, in these cells. Importantly, we have confirmed that such upregulated gene expression results in protein production with the demonstration that S. aureus challenge elicits the rapid and robust release of these chemokines by osteoblasts and does so in a bacterial dose-dependent manner. Furthermore, we have confirmed the ability of soluble osteoblast-derived chemokines to elicit the migration of a neutrophil-like cell line. As such, these studies demonstrate the robust production of CXCL1, CXCL2, CXCL3, CXCL5, CCL3, and CCL7 by osteoblasts in response to S. aureus infection, and the release of such neutrophil-attracting chemokines provides an additional mechanism by which osteoblasts could drive the inflammatory bone loss associated with staphylococcal osteomyelitis.


Asunto(s)
Osteomielitis , Infecciones Estafilocócicas , Animales , Ratones , Staphylococcus aureus/metabolismo , Neutrófilos/metabolismo , Quimiocinas/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Osteoblastos , Interleucina-8/metabolismo , Infecciones Estafilocócicas/microbiología , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Quimiocina CCL7/metabolismo , Quimiocina CCL3/metabolismo
3.
Mol Pain ; 19: 17448069231169373, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36998150

RESUMEN

BACKGROUND: Chemokine-mediated neuroinflammation plays an important role in the pathogenesis of neuropathic pain. The chemokine CC motif ligand 7 (CCL7) and its receptor CCR2 have been reported to contribute to neuropathic pain via astrocyte-microglial interaction in the spinal cord. Whether CCL7 in the trigeminal ganglion (TG) involves in trigeminal neuropathic pain and the involved mechanism remain largely unknown. METHODS: The partial infraorbital nerve transection (pIONT) was used to induce trigeminal neuropathic pain in mice. The expression of Ccl7, Ccr1, Ccr2, and Ccr3 was examined by real-time quantitative polymerase chain reaction. The distribution of CCL7, CCR2, and CCR3 was detected by immunofluorescence double-staining. The activation of extracellular signal-regulated kinase (ERK) was examined by Western blot and immunofluorescence. The effect of CCL7 on neuronal excitability was tested by whole-cell patch clamp recording. The effect of selective antagonists for CCR1, CCR2, and CCR3 on pain hypersensitivity was checked by behavioral testing. RESULTS: Ccl7 was persistently increased in neurons of TG after pIONT, and specific inhibition of CCL7 in the TG effectively relieved pIONT-induced orofacial mechanical allodynia. Intra-TG injection of recombinant CCL7 induced mechanical allodynia and increased the phosphorylation of ERK in the TG. Incubation of CCL7 with TG neurons also dose-dependently enhanced the neuronal excitability. Furthermore, pIONT increased the expression of CCL7 receptors Ccr1, Ccr2, and Ccr3. The intra-TG injection of the specific antagonist of CCR2 or CCR3 but not of CCR1 alleviated pIONT-induced orofacial mechanical allodynia and reduced ERK activation. Immunostaining showed that CCR2 and CCR3 are expressed in TG neurons, and CCL7-induced hyperexcitability of TG neurons was decreased by antagonists of CCR2 or CCR3. CONCLUSION: CCL7 activates ERK in TG neurons via CCR2 and CCR3 to enhance neuronal excitability, which contributes to the maintenance of trigeminal neuropathic pain. CCL7-CCR2/CCR3-ERK pathway may be potential targets for treating trigeminal neuropathic pain.


Asunto(s)
Quimiocina CCL7 , Quinasas MAP Reguladas por Señal Extracelular , Neuralgia , Neuralgia del Trigémino , Animales , Ratones , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Quimiocina CCL7/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hiperalgesia/metabolismo , Ligandos , Sistema de Señalización de MAP Quinasas , Neuralgia/metabolismo , Ganglio del Trigémino/metabolismo , Neuralgia del Trigémino/metabolismo , Receptores CCR2/metabolismo , Receptores CCR3/metabolismo
4.
Sci Rep ; 12(1): 19026, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36347994

RESUMEN

Kruppel like factor 15 (KLF15), a transcriptional factor belonging to the Kruppel-like factor (KLF) family of genes, has recently been reported as a tumor suppressor gene in breast cancer. However, the specific mechanisms by which KLF15 inhibits BrCa have not been elucidated. Here we investigated the role and mechanism of KLF15 in triple-negative breast cancer (TNBC). KLF15 expression and methylation were detected by RT-qPCR, RT-PCR and methylation-specific PCR in breast cancer cell lines and tissues. The effects of KLF15 on TNBC cell functions were examined via various cellular function assays. The specific anti-tumor mechanisms of KLF15 were further investigated by RNA sequence, RT-qPCR, Western blotting, luciferase assay, ChIP, and bioinformatics analysis. As the results showed that KLF15 is significantly downregulated in breast cancer cell lines and tissues, which promoter methylation of KLF15 partially contributes to. Exogenous expression of KLF15 induced apoptosis and G2/M phase cell cycle arrest, suppressed cell proliferation, metastasis and in vivo tumorigenesis of TNBC cells. Mechanism studies revealed that KLF15 targeted and downregulated C-C motif chemokine ligand 2 (CCL2) and CCL7. Moreover, transcriptome and metabolome analysis revealed that KLF15 is involved in key anti-tumor regulatory and metabolic pathways in TNBC. In conclusion, KLF15 suppresses cell growth and metastasis in TNBC by downregulating CCL2 and CCL7. KLF15 may be a prognostic biomarker in TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Ligandos , Proliferación Celular/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Quimiocinas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Quimiocina CCL7/metabolismo , Quimiocina CCL2/metabolismo
5.
Science ; 378(6621): eabl7207, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36395212

RESUMEN

Many human cancers manifest the capability to circumvent attack by the adaptive immune system. In this work, we identified a component of immune evasion that involves frequent up-regulation of fragile X mental retardation protein (FMRP) in solid tumors. FMRP represses immune attack, as revealed by cancer cells engineered to lack its expression. FMRP-deficient tumors were infiltrated by activated T cells that impaired tumor growth and enhanced survival in mice. Mechanistically, FMRP's immunosuppression was multifactorial, involving repression of the chemoattractant C-C motif chemokine ligand 7 (CCL7) concomitant with up-regulation of three immunomodulators-interleukin-33 (IL-33), tumor-secreted protein S (PROS1), and extracellular vesicles. Gene signatures associate FMRP's cancer network with poor prognosis and response to therapy in cancer patients. Collectively, FMRP is implicated as a regulator that orchestrates a multifaceted barrier to antitumor immune responses.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Evasión Inmune , Tolerancia Inmunológica , Neoplasias , Animales , Humanos , Ratones , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Neoplasias/inmunología , Quimiocina CCL7/metabolismo , Interleucina-33 , Proteína S/metabolismo
6.
Int Immunopharmacol ; 113(Pt A): 109332, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36274485

RESUMEN

Natural killer (NK) cell-based therapy has been studied for the treatment of patients with cancers, but the inadequate infiltration of NK cells into solid tumors remains a big challenge to its clinical application. In this study, we examined the interaction between NK cells and endothelial cells, which might play a major role in NK cell homing to solid tumors. We found that endothelial cells were activated by TNF-α and IL-1ß, which were produced by tumor-associated CD11b+ cells, which included F4/80+ macrophages. TNF-α-treated endothelial cells increased NK cell migration by producing CCL2 and CCL7, which was proved by transwell and imaging assays. TNF-α-treated endothelial cells adhered well to NK cells, which was due to a TNF-α-induced increase in ICAM-1 and VCAM-1 expression on endothelial cells. Imaging data confirmed that TNF-α-treated endothelial cells transfected with ICAM-1 or VCAM-1 siRNAs did not establish stable contacts with NK cells. Taken together, our data suggest that CCL2, CCL7, ICAM-1, and VCAM-1 expressed by endothelial cells will be potential targets to guide adequate interaction with NK cells, which is a crucial step for NK cell homing to the tumor microenvironment.


Asunto(s)
Molécula 1 de Adhesión Intercelular , Molécula 1 de Adhesión Celular Vascular , Humanos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Células Endoteliales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Endotelio Vascular/metabolismo , Células Asesinas Naturales/metabolismo , Células Cultivadas , Quimiocina CCL7/metabolismo , Quimiocina CCL2/metabolismo
7.
Cardiovasc Diabetol ; 21(1): 185, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109744

RESUMEN

Chemokines are key components in the pathology of chronic diseases. Chemokine CC motif ligand 7 (CCL7) is believed to be associated with cardiovascular disease, diabetes mellitus, and kidney disease. CCL7 may play a role in inflammatory events by attracting macrophages and monocytes to further amplify inflammatory processes and contribute to disease progression. However, CCL7-specific pathological signaling pathways need to be further confirmed in these chronic diseases. Given the multiple redundancy system among chemokines and their receptors, further experimental and clinical studies are needed to clarify whether direct CCL7 inhibition mechanisms could be a promising therapeutic approach to attenuating the development of cardiovascular disease, diabetes mellitus, and kidney disease.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus , Enfermedades Renales , Quimiocina CCL7/metabolismo , Quimiocinas/metabolismo , Humanos , Ligandos
8.
Biomed Pharmacother ; 153: 113549, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076613

RESUMEN

Microglial activation in the spinal cord contributes to the development of inflammatory pain. Monocyte chemotactic protein 3 (MCP3) can induce microglial activation, resulting in increased pain sensitivity; however, the underlying mechanism remains poorly understood. 3,5-dicaffeoylquinic acid (3,5-DCQA) has shown protective effects against inflammation-related diseases, but the effect of 3,5-DCQA on microglial activation and inflammatory pain is not evaluated. This study aimed to investigate the effects of 3,5-DCQA on microglial activation-induced inflammatory pain. Furthermore, the underlying mechanism inhibited by 3,5-DCQA via MCP3 suppression was studied. To induce microglial activation, LPS was treated in BV2 microglial cells. The LPS-induced microglial activation and pro-inflammatory cytokines production were significantly reduced by 3,5-DCQA treatment in BV2 cells. Moreover, 3,5-DCQA suppressed LPS-induced MCP3 expression, resulting in reduced phosphorylation of JAK2/STAT3. Interestingly, the suppressed JAK2/STAT3 signaling enhanced autophagy induction in BV2 cells. The increased autophagy by 3,5-DCQA and knockout of MCP3 inhibited LPS-induced inflammatory response in BV2 cells. To establish the inflammatory pain, CFA was injected into the right paw of mice. The CFA-induced pain hypersensitivity and foot swelling were attenuated by the oral administration of 3,5-DCQA. Moreover, CFA-induced microglial activation was reduced and the autophagy markers were recovered in the spinal cord of 3,5-DCQA-administered mice. Similar results were observed in cultured primary microglia. Our findings indicate that 3,5-DCQA attenuates inflammation-mediated pain hypersensitivity by enhancing autophagy through inhibition of MCP3-induced JAK2/STAT3 signaling. Therefore, 3,5-DCQA could be a potential therapeutic agent for alleviating inflammatory pain.


Asunto(s)
Ácido Clorogénico , Lipopolisacáridos , Microglía , Animales , Autofagia/efectos de los fármacos , Quimiocina CCL7/efectos de los fármacos , Quimiocina CCL7/metabolismo , Ácido Clorogénico/análogos & derivados , Ácido Clorogénico/farmacología , Inflamación/metabolismo , Janus Quinasa 2/efectos de los fármacos , Janus Quinasa 2/metabolismo , Lipopolisacáridos/farmacología , Ratones , Dolor/tratamiento farmacológico , Dolor/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo
9.
J Immunol Res ; 2022: 6450721, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36118415

RESUMEN

Objective: Infiltration of tumor-associated macrophages is closely linked to the malignant development of human cancers. This research studies the function of C-C motif chemokine ligand 7 (CCL7) in the macrophage accumulation in lung adenocarcinoma (LUAD) and the underpinning mechanism. Methods: The expression profile of CCL7 in LUAD and its correlations with patient's prognosis and macrophage infiltration were predicted via bioinformatics systems. Artificial up- or downregulation of CCL7 was induced in LUAD cells to explore its function in the mobility, EMT of cancer cells, and migration of M2 macrophages. Cancer cells were implanted in NOD/SCID mice to induce xenograft tumors. The CCL7-related transcription factors or factors were predicted by bioinformatic tools, and the molecular interactions were confirmed by immunoprecipitation or luciferase assays. Results: CCL7 was highly expressed in LUAD and linked to increased TAM infiltration. Knockdown of CCL7 suppressed the chemotaxis and M2 skewing of macrophages, and it blocked the EMT and mobility of LUAD cells. CCL7 downregulation also suppressed macrophage infiltration in xenograft tumors in mice. Spi-1 proto-oncogene (SPI1) was confirmed as an upstream factor activating CCL7 transcription, and LINC01094 was found to bind to SPI1 to promote its nuclear translocation. Upregulation of SPI1 restored the chemotactic migration and M2 polarization of macrophages in LUAD cells. Conclusion: This paper reveals that LINC01094 binds to SPI1 to promote its nuclear translocation, which further activates CCL7 transcription by binding to its promoter, leading to M2 macrophage accumulation and dissemination of tumor cells.


Asunto(s)
Adenocarcinoma del Pulmón , Quimiocina CCL7/metabolismo , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas/metabolismo , ARN Largo no Codificante/metabolismo , Transactivadores/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Quimiocinas/metabolismo , Humanos , Ligandos , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Cell Commun Signal ; 20(1): 94, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715847

RESUMEN

BACKGROUND: Chemoattractant is critical to recruitment of osteoclast precursors and stimulates tumor bone metastasis. However, the role of chemoattractant in bone metastasis of colorectal cancer (CRC) is still unclear. METHODS: Histochemistry analysis and TRAP staining were utilized to detect the bone resorption and activation of osteoclasts (OCs) after administration of CCL7 neutralizing antibody or CCR1 siRNA. qRT-PCR analysis and ELISA assay were performed to detect the mRNA level and protein level of chemoattractant. BrdU assay and Tunel assay were used to detect the proliferation and apoptosis of osteoclast precursors (OCPs). The migration of OCPs was detected by Transwell assay. Western blots assay was performed to examine the protein levels of pathways regulating the expression of CCL7 or CCR1. RESULTS: OCPs-derived CCL7 was significantly upregulated in bone marrow after bone metastasis of CRC. Blockage of CCL7 efficiently prevented bone resorption. Administration of CCL7 promoted the migration of OCPs. Lactate promoted the expression of CCL7 through JNK pathway. In addition, CCR1 was the most important receptor of CCL7. CONCLUSION: Our study indicates the essential role of CCL7-CCR1 signaling for recruitment of OCPs in early bone metastasis of CRC. Targeting CCL7 or CCR1 could restore the bone volume, which could be a potential therapeutical target. Video Abstract.


Asunto(s)
Neoplasias Óseas , Quimiocina CCL7 , Neoplasias Colorrectales , Osteoclastos , Osteólisis , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Huesos/metabolismo , Huesos/patología , Quimiocina CCL7/metabolismo , Factores Quimiotácticos/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Osteoclastos/patología , Osteólisis/metabolismo , Regulación hacia Arriba
11.
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
12.
Chem Biol Interact ; 355: 109804, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-35123994

RESUMEN

Recent reports have challenged the notion that the lens is immune-privileged. However, these studies have not fully identified the molecular mechanism(s) that promote immune surveillance of the lens. Using a mouse model of targeted glutathione (GSH) deficiency in ocular surface tissues, we have investigated the role of oxidative stress in upregulating cytokine expression and promoting immune surveillance of the eye. RNA-sequencing of lenses from postnatal day (P) 1-aged Gclcf/f;Le-CreTg/- (KO) and Gclcf/f;Le-Cre-/- control (CON) mice revealed upregulation of many cytokines (e.g., CCL4, GDF15, CSF1) and immune response genes in the lenses of KO mice. The eyes of KO mice had a greater number of cells in the aqueous and vitreous humors at P1, P20 and P50 than age-matched CON and Gclcw/w;Le-CreTg/- (CRE) mice. Histological analyses revealed the presence of innate immune cells (i.e., macrophages, leukocytes) in ocular structures of the KO mice. At P20, the expression of cytokines and ROS content was higher in the lenses of KO mice than in those from age-matched CRE and CON mice, suggesting that oxidative stress may induce cytokine expression. In vitro administration of the oxidant, hydrogen peroxide, and the depletion of GSH (using buthionine sulfoximine (BSO)) in 21EM15 lens epithelial cells induced cytokine expression, an effect that was prevented by co-treatment of the cells with N-acetyl-l-cysteine (NAC), a antioxidant. The in vivo and ex vivo induction of cytokine expression by oxidative stress was associated with the expression of markers of epithelial-to-mesenchymal transition (EMT), α-SMA, in lens cells. Given that EMT of lens epithelial cells causes posterior capsule opacification (PCO), we propose that oxidative stress induces cytokine expression, EMT and the development of PCO in a positive feedback loop. Collectively these data indicate that oxidative stress induces inflammation of lens cells which promotes immune surveillance of ocular structures.


Asunto(s)
Ojo/anatomía & histología , Inmunidad Innata , Cristalino/metabolismo , Estrés Oxidativo , Acetilcisteína/farmacología , Animales , Butionina Sulfoximina/farmacología , Línea Celular , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Ojo/metabolismo , Glutamato-Cisteína Ligasa/deficiencia , Glutamato-Cisteína Ligasa/genética , Cristalino/citología , Leucocitos/citología , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos
13.
Front Immunol ; 13: 993444, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36685592

RESUMEN

Glioblastoma (GBM) is the most common and malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized in part by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive, hematopoietic cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a potent subset of myeloid cells, expressing monocytic (M)-MDSC markers, distinguished by dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate into the TME. This study evaluated the T cell suppressive function and migratory properties of CCR2+/CX3CR1+ MDSCs. Bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Recombinant and glioma-derived CCL2 and CCL7 induce the migration of CCR2+/CX3CR1+ MDSCs with similar efficacy. KR158B-CCL2 and -CCL7 knockdown murine gliomas contain equivalent percentages of CCR2+/CX3CR1+ MDSCs compared to KR158B gliomas. Combined neutralization of CCL2 and CCL7 completely blocks CCR2-expressing cell migration to KR158B cell conditioned media. CCR2+/CX3CR1+ cells are also reduced within KR158B gliomas upon combination targeting of CCL2 and CCL7. High levels of CCL2 and CCL7 are also associated with negative prognostic outcomes in GBM patients. These data provide a more comprehensive understanding of the function of CCR2+/CX3CR1+ MDSCs and the role of CCL2 and CCL7 in the recruitment of these immune suppressive cells and further support the significance of targeting this chemokine axis in GBM.


Asunto(s)
Glioblastoma , Glioma , Células Supresoras de Origen Mieloide , Animales , Ratones , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Receptor 1 de Quimiocinas CX3C/metabolismo , Glioblastoma/patología , Monocitos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Microambiente Tumoral
14.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948231

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease with an unmet need of biomarkers that can aid in the diagnostic and prognostic assessment of the disease and response to treatment. In this two-part explorative proteomic study, we demonstrate how proteins associated with tissue remodeling, inflammation and chemotaxis such as MMP7, CXCL13 and CCL19 are released in response to aberrant extracellular matrix (ECM) in IPF lung. We used a novel ex vivo model where decellularized lung tissue from IPF patients and healthy donors were repopulated with healthy fibroblasts to monitor locally released mediators. Results were validated in longitudinally collected serum samples from 38 IPF patients and from 77 healthy controls. We demonstrate how proteins elevated in the ex vivo model (e.g., MMP7), and other serum proteins found elevated in IPF patients such as HGF, VEGFA, MCP-3, IL-6 and TNFRSF12A, are associated with disease severity and progression and their response to antifibrotic treatment. Our study supports the model's applicability in studying mechanisms involved in IPF and provides additional evidence for both established and potentially new biomarkers in IPF.


Asunto(s)
Biomarcadores/metabolismo , Microambiente Celular/fisiología , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Anciano , Quimiocina CCL7/metabolismo , Quimiocina CXCL13/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Masculino , Metaloproteinasa 7 de la Matriz/metabolismo , Persona de Mediana Edad , Proteómica/métodos , Receptor de TWEAK/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Physiol Rep ; 9(19): e14997, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34605213

RESUMEN

Acrolein is a reactive inhalation hazard. Acrolein's initial interaction, which in itself can be function-altering, is followed by time-dependent cascade of complex cellular and pulmonary responses that dictate the severity of the injury. To investigate the pathophysiological progression of sex-dependent acrolein-induced acute lung injury, C57BL/6J mice were exposed for 30 min to sublethal, but toxic, and lethal acrolein. Male mice were more sensitive than female mice. Acrolein of 50 ppm was sublethal to female but lethal to male mice, and 75 ppm was lethal to female mice. Lethal and sublethal acrolein exposure decreased bronchoalveolar lavage (BAL) total cell number at 3 h after exposure. The cell number decrease was followed by progressive total cell and neutrophil number and protein increases. The BAL total cell number in female mice exposed to a sublethal, but not lethal dose, returned to control levels at 16 h. In contrast, BAL protein content and neutrophil number were higher in mice exposed to lethal compared to sublethal acrolein. RNASeq pathway analysis identified greater increased lung neutrophil, glutathione metabolism, oxidative stress responses, and CCL7 (aka MCP-3), CXCL10 (aka IP-10), and IL6 transcripts in males than females, whereas IL10 increased more in female than male mice. Thus, the IL6:IL10 ratio, an indicator of disease severity, was greater in males than females. Further, H3.3 histone B (H3F3B) and pro-platelet basic protein (PPBP aka CXCL7), transcripts increased in acrolein exposed mouse BAL and plasma at 3 h, while H3F3B protein that is associated with neutrophil extracellular traps formation increased at 12 h. These results suggest that H3F3B and PPBP transcripts increase may contribute to extracellular H3F3B and PPBP proteins increase.


Asunto(s)
Acroleína/toxicidad , Lesión Pulmonar Aguda/inducido químicamente , Pulmón/efectos de los fármacos , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Animales , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Femenino , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Factores Sexuales
16.
Redox Biol ; 46: 102079, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34454163

RESUMEN

Liver injuries induced by various stimuli share in common an acute inflammatory response, in which circulating macrophages home to the liver parenchyma to participate in the regulation of repair, regeneration, and fibrosis. In the present study we investigated the role of hepatocyte-derived C-C motif ligand 7 (CCL7) in macrophage migration during liver injury focusing on its transcriptional regulation. We report that CCL7 expression was up-regulated in the liver by lipopolysaccharide (LPS) injection (acute liver injury) or methionine-and-choline-deficient (MCD) diet feeding (chronic liver injury) paralleling increased macrophage infiltration. CCL7 expression was also inducible in hepatocytes, but not in hepatic stellate cells or in Kupffer cells, by LPS treatment or exposure to palmitate in vitro. Hepatocyte-specific deletion of Brahma-related gene 1 (BRG1), a chromatin remodeling protein, resulted in a concomitant loss of CCL7 induction and macrophage infiltration in the murine livers. Of interest, BRG1-induced CCL7 transcription and macrophage migration was completely blocked by the antioxidant N-acetylcystine. Further analyses revealed that BRG1 interacted with activator protein 1 (AP-1) to regulate CCL7 transcription in hepatocytes in a redox-sensitive manner mediated in part by casein kinase 2 (CK2)-catalyzed phosphorylation of BRG1. Importantly, a positive correlation between BRG1/CCL7 expression and macrophage infiltration was identified in human liver biopsy specimens. In conclusion, our data unveil a novel role for BRG1 as a redox-sensitive activator of CCL7 transcription.


Asunto(s)
ADN Helicasas , Proteínas Nucleares , Animales , Células Cultivadas , Quimiocina CCL7/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Factores de Transcripción
17.
Int Immunopharmacol ; 99: 107975, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34293712

RESUMEN

Microglia are immune cells of the central nervous system that mediate neuroinflammation. It is widely known that microglia-mediated inflammation in the brain contribute to the widespread tissue damage and neurological deficits in traumatic brain injury (TBI). However, the mechanisms responsible for this inflammatory response remain elusive. Here, we investigated the role of astrocyte-derived chemokine (C-C motif) ligand 7 (CCL7) in microglial-controlled inflammation following TBI. Our results demonstrated that astrocyte-derived CCL7 induced microglial activation and the release of proinflammatory mediators in the cortex and serum of rats that underwent experimental TBI. Furthermore, CCL7 knockout improved microglia-controlled inflammation, brain morphology and neurological dysfunction following TBI. In vitro, CCL7-siRNA attenuated the LPS-induced expression of pro-inflammatory markers in the co-culture of microglia and astrocytes. Collectively, our findings uncover an important role for astrocyte-derived CCL7 in promoting microglia-mediated inflammation after TBI and suggests CCL7 could serve as a potential therapeutic strategy for attenuating TBI by inhibiting microglial activation.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Quimiocina CCL7/farmacología , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/patología , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Quimiocina CCL7/metabolismo , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Enfermedades Neuroinflamatorias/psicología , Cultivo Primario de Células , ARN Interferente Pequeño/farmacología , Reconocimiento en Psicología/efectos de los fármacos
18.
J Cell Mol Med ; 25(15): 7280-7293, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34189838

RESUMEN

Chemokine C-C motif ligand 7 (CCL7), a member of CC chemokine subfamily, plays pivotal roles in numerous inflammatory diseases. Hyper-activation of inflammation is an important characteristic of abdominal aortic aneurysm (AAA). Therefore, in the present study, we aimed to determine the effect of CCL7 on AAA formation. CCL7 abundance in aortic tissue and macrophage infiltration were both increased in angiotensin II (Ang II)-induced AAA mice. Ex vivo, CCL7 promoted macrophage polarization towards M1 phenotype. This effect was reversed by the blockage of CCR1, a receptor of CCL7. CCL7 up-regulated JAK2/STAT1 protein level in macrophage, and CCL7-induced M1 activation was suppressed by JAK2/STAT1 pathway inhibition. To verify the effect of CCL7 on AAA in vivo, either CCL7-neutralizing antibody (CCL7-nAb) or vehicles were intraperitoneally injected 24 hours prior to Ang II infusion and subsequently every three days for 4 weeks. CCL7-nAb administration significantly attenuated Ang II-induced luminal and external dilation as well as pathological remodelling. Immunostaining showed that CCL7-nAb administration significantly decreased aneurysmal macrophage infiltration. In conclusion, CCL7 contributed to Ang II-induced AAA by promoting M1 phenotype of macrophage through CCR1/JAK2/STAT1 signalling pathway.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Movimiento Celular , Quimiocina CCL7/metabolismo , Macrófagos/metabolismo , Angiotensina II/toxicidad , Animales , Aneurisma de la Aorta Abdominal/etiología , Aneurisma de la Aorta Abdominal/patología , Diferenciación Celular , Células Cultivadas , Quimiocina CCL7/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Receptores CCR1/metabolismo , Factor de Transcripción STAT1/metabolismo , Remodelación Vascular
19.
BMC Immunol ; 22(1): 27, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849432

RESUMEN

BACKGROUND: Food proteins differ in their allergenic potential. Currently, there is no predictive and validated bio-assay to evaluate the allergenicity of novel food proteins. The objective of this study was to investigate the potential of a human peripheral blood mononuclear cell (PBMC) gene expression assay to identify biomarkers to predict the allergenicity of legume proteins. RESULTS: PBMCs from healthy donors were exposed to weakly and strongly allergenic legume proteins (2S albumins, and 7S and 11S globulins from white bean, soybean, peanut, pea and lupine) in three experiments. Possible biomarkers for allergenicity were investigated by exposing PBMCs to a protein pair of weakly (white bean) and strongly allergenic (soybean) 7S globulins in a pilot experiment. Gene expression was measured by RNA-sequencing and differentially expressed genes were selected as biomarkers. 153 genes were identified as having significantly different expression levels to the 7S globulin of white bean compared to soybean. Inclusion of multiple protein pairs from 2S albumins (lupine and peanut) and 7S globulins (white bean and soybean) in a larger study, led to the selection of CCL2, CCL7, and RASD2 as biomarkers to distinguish weakly from strongly allergenic proteins. The relevance of these three biomarkers was confirmed by qPCR when PBMCs were exposed to a larger panel of weakly and strongly allergenic legume proteins (2S albumins, and 7S and 11S globulins from white bean, soybean, peanut, pea and lupine). CONCLUSIONS: The PBMC gene expression assay can potentially distinguish weakly from strongly allergenic legume proteins within a protein family, though it will be challenging to develop a generic method for all protein families from plant and animal sources. Graded responses within a protein family might be of more value in allergenicity prediction instead of a yes or no classification.


Asunto(s)
Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Hipersensibilidad a los Alimentos/inmunología , Proteínas de Unión al GTP/metabolismo , Leucocitos Mononucleares/fisiología , Albuminas 2S de Plantas/inmunología , Alérgenos/inmunología , Antígenos de Plantas/inmunología , Biomarcadores/metabolismo , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL7/genética , Fabaceae/inmunología , Proteínas de Unión al GTP/genética , Globulinas/inmunología , Humanos , Inmunoglobulina E/metabolismo , Proteínas de Almacenamiento de Semillas/inmunología , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad , Proteínas de Soja/inmunología , Transcriptoma
20.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671469

RESUMEN

Bone metastasis remains the most frequent and the deadliest complication of prostate cancer (PCa). Mechanisms leading to the homing of tumor cells to bone remain poorly characterized. Role of chemokines in providing navigational cues to migrating cancer cells bearing specific receptors is well established. Bone is an adipocyte-rich organ since 50 to 70% of the adult bone marrow (BM) volume comprise bone marrow adipocytes (BM-Ads), which are likely to produce chemokines within the bone microenvironment. Using in vitro migration assays, we demonstrated that soluble factors released by human primary BM-Ads are able to support the directed migration of PCa cells in a CCR3-dependent manner. In addition, we showed that CCL7, a chemokine previously involved in the CCR3-dependent migration of PCa cells outside of the prostate gland, is released by human BM-Ads. These effects are amplified by obesity and ageing, two clinical conditions known to promote aggressive and metastatic PCa. In human tumors, we found an enrichment of CCR3 in bone metastasis vs. primary tumors at mRNA levels using Oncomine microarray database. In addition, immunohistochemistry experiments demonstrated overexpression of CCR3 in bone versus visceral metastases. These results underline the potential importance of BM-Ads in the bone metastatic process and imply a CCR3/CCL7 axis whose pharmacological interest needs to be evaluated.


Asunto(s)
Adipocitos/metabolismo , Adipocitos/patología , Médula Ósea/patología , Huesos/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores CCR3/metabolismo , Envejecimiento/patología , Médula Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Línea Celular Tumoral , Quimiocina CCL7/metabolismo , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Masculino , Metástasis de la Neoplasia , Obesidad/complicaciones , Neoplasias de la Próstata/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...