Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 516
Filtrar
1.
Tissue Cell ; 86: 102294, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38181585

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic joint inflammation. Fibronectin type III domain-containing protein 4 (FNDC4) is a secretory factor that can regulate inflammatory diseases. However, the role of FNDC4 in RA has not been reported so far. METHODS: The expression of FNDC4 in synovial tissues of RA was analyzed by GEO database (GSE55235 dataset). Then, the expression of FNDC4 in RA fibroblast-like synoviocytes (RA-FLSs) was detected by RT-qPCR and western blot. After constructing FNDC4 overexpression plasmid, cell proliferation and apoptosis were detected. Wound healing and transwell assays were used to detect cell migration and invasion. Then we examined the expression of cytokines related to cell inflammation. Subsequently, the regulatory mechanism of FNDC4 was further discussed. We detected the expression of CCL2 and ERK signaling pathway related proteins downstream of FNDC4. Finally, the mechanism was discussed through the overexpression of FNDC4 and CCL2 and the addition of ERK pathway activator tBHQ. RESULTS: GEO database showed that FNDC4 expression decreased in synovial tissues of RA. FNDC4 expression was also decreased in RA-FLSs. Overexpression of FNDC4 inhibited the proliferation, invasion and migration of RA-FLSs whereas promoted the cellapoptosis. Overexpression of FNDC4 inhibited the release of inflammatory factors in RA-FLSs. The regulatory effect of FNDC4 is achieved by inhibiting the CCL2/ERK signaling pathway. CONCLUSION: FNDC4 reduces inflammation, proliferation, invasion and migration of RA-FLSs in RA by inhibiting CCL2/ERK signaling.


Asunto(s)
Artritis Reumatoide , Sinoviocitos , Humanos , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Fibroblastos/metabolismo , Inflamación/genética , Inflamación/metabolismo , Transducción de Señal/genética , Membrana Sinovial , Sinoviocitos/metabolismo , Fibronectinas/metabolismo
2.
J Control Release ; 365: 358-368, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38016488

RESUMEN

Blood vessel functionality is crucial for efficient tumor-targeted drug delivery. Heterogeneous distribution and perfusion of angiogenic blood vessels contribute to suboptimal accumulation of (nano-) therapeutics in tumors and metastases. To attenuate pathological angiogenesis, an L-RNA aptamer inhibiting the CC motif chemokine ligand 2 (CCL2) was administered to mice bearing orthotopic 4T1 triple-negative breast cancer tumors. The effect of CCL2 inhibition on tumor blood vessel functionality and tumor-targeted drug delivery was evaluated via multimodal and multiscale optical imaging, employing fluorophore-labeled polymeric (10 nm) and liposomal (100 nm) nanocarriers. Anti-CCL2 treatment induced a dose-dependent anti-angiogenic effect, reflected by a decreased relative blood volume, increased blood vessel maturity and functionality, and reduced macrophage infiltration, accompanied by a shift in the polarization of tumor-associated macrophages (TAM) towards a less M2-like and more M1-like phenotype. In line with this, CCL2 inhibitor treatment improved the delivery of polymers and liposomes to tumors, and enhanced the antitumor efficacy of free and liposomal doxorubicin. Together, these findings demonstrate that blocking the CCL2-CCR2 axis modulates TAM infiltration and polarization, resulting in vascular normalization and improved tumor-targeted drug delivery.


Asunto(s)
Quimiocina CCL2 , Neoplasias , Ratones , Animales , Quimiocina CCL2/farmacología , Ligandos , Nanomedicina , Neoplasias/patología , Macrófagos , Línea Celular Tumoral
3.
Mol Neurobiol ; 61(3): 1433-1447, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37721689

RESUMEN

Spinal cord injury (SCI) is a common clinical problem in orthopedics with a lack of effective treatments and drug targets. In the present study, we performed bioinformatic analysis of SCI datasets GSE464 and GSE45006 in the Gene Expression Omnibus (GEO) public database and experimentally validated CCL2 expression in an animal model of SCI. This was followed by stimulation of PC-12 cells using hydrogen peroxide to construct a cellular model of SCI. CCL2 expression was knocked down using small interfering RNA (si-CCL2), and PI3K signaling pathway inhibitors and activators were used to validate and observe the changes in downstream inflammation. Through data mining, we found that the inflammatory chemokine CCL2 and PI3K/Akt signaling pathways after SCI expression were significantly increased, and after peroxide stimulation of PC-12 cells with CCL2 knockdown, their downstream cellular inflammatory factor levels were decreased. The PI3K/Akt signaling pathway was blocked by PI3K inhibitors, and the downstream inflammatory response was suppressed. In contrast, when PI3K activators were used, the inflammatory response was enhanced, indicating that the CCL2-PI3K/Akt signaling pathway plays a key role in the regulation of the inflammatory response. This study revealed that the inflammatory chemokine CCL2 can regulate the inflammatory response of PC-12 cells through the PI3K/Akt signaling pathway, and blocking the expression of the inflammatory chemokine CCL2 may be a promising strategy for the treatment of secondary injury after SCI.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Traumatismos de la Médula Espinal , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Quimiocina CCL2/farmacología , Transducción de Señal , Traumatismos de la Médula Espinal/metabolismo , Biología Computacional , Médula Espinal/metabolismo
4.
Cornea ; 42(8): 1005-1015, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37155343

RESUMEN

PURPOSE: The aim of this study was to investigate the effects of indoleamine 2,3-dioxygenase (IDO) on macrophage polarization, phagocytosis, and killing through regulation of the CCL2/CCR2 signaling pathway in Aspergillus fumigatus keratitis. METHODS: In vivo and in vitro experiments were conducted in mice and mouse peritoneal macrophages after infection with A. fumigatus . Clinical scoring, reverse transcription-polymerase chain reaction, and immunofluorescence staining were used to evaluate the fungal keratitis lesions, macrophage-related cytokines, and macrophage recruitment. The expression of CCL2 and CCR2 was detected by reverse transcription-polymerase chain reaction and western blot after pretreatment with or without an IDO inhibitor (1-MT). After pretreatment with 1-MT, a CCR2 antagonist, a CCL2 neutralizing antibody, an IDO agonist (IFNG), and recombinant CCL2 protein (CCL2), the flow cytometry and colony-forming unit counts were used to detect the polarization, phagocytosis, and killing function. RESULTS: Compared with the control group, the infected eyes showed increased clinical scores, macrophage-related cytokine expression, and macrophage recruitment. 1-MT pretreatment increased the expression of CCL2 and CCR2 and the proportion of CD206+/CD86+ macrophages; macrophages polarized toward the M2 type, with enhanced killing function. CCR2 antagonists and CCL2 neutralizing antibodies reversed the effects of 1-MT. Compared with the infected group, IFNG pretreatment decreased the proportion of CD206+/CD86+ macrophages, and macrophages polarized toward the M1 type, with decreased phagocytosis and impaired killing function. CCL2 reversed the effect of IFNG. CONCLUSIONS: IDO can promote the polarization of macrophages to the M1 type by blocking the CCL2/CCR2 signaling pathway, inhibiting the phagocytosis and killing function of macrophages, and mediating the protective immune role of A. fumigatus .


Asunto(s)
Úlcera de la Córnea , Queratitis , Animales , Ratones , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Úlcera de la Córnea/metabolismo , Citocinas/metabolismo , Queratitis/patología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Proteínas Recombinantes , Transducción de Señal , Indolamina-Pirrol 2,3,-Dioxigenasa
5.
Metab Brain Dis ; 38(4): 1335-1349, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36795287

RESUMEN

Spinal cord injury (SCI) is a devastating neurological disease with no cure that usually results in irreversible loss of sensory and voluntary motor functions below the injury site. We conducted an in-depth bioinformatics analysis combining the gene expression omnibus spinal cord injury database and the autophagy database and found that the expression of the autophagy gene CCL2 was significantly upregulated and the PI3K/Akt/mTOR signaling pathway was activated after SCI. The results of the bioinformatics analysis were verified by constructing animal and cellular models of SCI. We then used small interfering RNA to inhibit the expression of CCL2 and PI3K to inhibit and activate the PI3K/Akt/mTOR signaling pathway; western blot, immunofluorescence, monodansylcadaverine, and cell flow techniques were used to detect the expression of key proteins involved in downstream autophagy and apoptosis. We found that when PI3K inhibitors were activated, apoptosis decreased, the levels of autophagy-positive proteins LC3-I/LC3-II and Bcl-1 increased, the levels of autophagy-negative protein P62 decreased, the levels of pro-apoptotic proteins Bax and caspase-3 decreased, the levels of the apoptosis-inhibiting protein Bcl-2 increased. In contrast, when a PI3K activator was used, autophagy was inhibited, and apoptosis was increased. This study revealed the effect of CCL2 on autophagy and apoptosis after SCI through the PI3K/Akt/mTOR signaling pathway. By blocking the expression of the autophagy-related gene CCL2, the autophagic protective response can be activated, and apoptosis can be inhibited, which may be a promising strategy for the treatment of SCI.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Traumatismos de la Médula Espinal , Ratas , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Apoptosis , Autofagia , Médula Espinal , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología
6.
Neurobiol Dis ; 174: 105892, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36240949

RESUMEN

Preventing brain cell loss and enhancing tissue repair are crucial objectives to improve the outcome of stroke. Fetal microchimerism has been implicated in brain repair following ischemic stroke in mice. CCL2/CCR2 signaling pathway triggers fetal progenitors trafficking to cutaneous wounds. Therefore, we sought to evaluate whether CCL2 could dampen brain damage in a model of excitotoxic lesion in post-partum mice. Virgin or post-partum mice were subjected to an intracerebral injection of ibotenate to induce excitotoxic lesions. Low doses of CCL2 or its vehicle were concomitantly injected. Morphological and molecular analyses were performed 1 and 5 days following the procedure. Intracerebral treatment with low doses of CCL2 was able to limit brain excitotoxic damage induced by ibotenate in post-partum mice, through an enhanced recruitment of fetal microchimeric cells to the damaged hemisphere. At day 1 post-injection, we observed a decreased cortical apoptosis associated with a reduced reactive astrocytosis. At day 5, we found an increased proportion of mature neurons and oligodendrocytes correlating with an increase in GAP43 growth cones. At this stage, immune microglial cells were reduced, while angiogenesis was enhanced. Importantly, CCL2 did not have beneficial effects in virgin mice therefore ruling out a specific role of CCL2 independently from fetal microchimeric cells mobilization. CCL2 treatment efficiently enhances fetal cell mobilization to improve the outcome of a brain excitotoxic challenge in post-partum mice. This study paves the way for a "natural stem cell therapy" based on the selective recruitment of fetal progenitors to repair maternal brain injury.


Asunto(s)
Lesiones Encefálicas , Humanos , Femenino , Animales , Ratones , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Periodo Posparto , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología
7.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35851310

RESUMEN

BACKGROUND: There was much hard work to study the trastuzumab resistance in HER2-positive gastric cancer (GC), but the information which would reveal this abstruse mechanism is little. In this study, we aimed to investigate the roles of tumor cell-derived CCL2 on trastuzumab resistance and overcome the resistance by treatment with the anti-CD40-scFv-linked anti-HER2 (CD40 ×HER2) bispecific antibody (bsAb). METHODS: We measured the levels of CCL2 expression in HER2-positive GC tissues, and revealed biological functions of tumor cell-derived CCL2 on tumor-associated macrophages (TAMs) and the trastuzumab resistance. Then, we developed CD40 ×HER2 bsAb, and examined the targeting roles on HER2 and CD40, to overcome the trastuzumab resistance without systemic toxicity. RESULTS: We found the level of CCL2 expression in HER2-postive GC was correlated with infiltration of TAMs, polarization status of infiltrated TAMs, trastuzumab resistance and survival outcomes of GC patients. On exposure to CCL2, TAMs decreased the M1-like phenotype, thereby eliciting the trastuzumab resistance. CCL2 activated the transcription of ZC3H12A, which increased K63-linked deubiquitination and K48-linked auto-ubiquitination of TRAF6/3 to inactivate NF-κB signaling in TAMs. CD40 ×HER2 bsAb, which targeted the CD40 to restore the ubiquitination level of TRAF6/3, increased the M1-like phenotypic transformation of TAMs, and overcame trastuzumab resistance without immune-related adversary effects (irAEs). CONCLUSIONS: We revealed a novel mechanism of trastuzumab resistance in HER2-positive GC via the CCL2-ZC3H12A-TRAF6/3 signaling axis, and presented a CD40 ×HER2 bsAb which showed great antitumor efficacy with few irAEs.


Asunto(s)
Neoplasias Gástricas , Antígenos CD40/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Humanos , Receptor ErbB-2/metabolismo , Transducción de Señal , Neoplasias Gástricas/patología , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/farmacología , Trastuzumab/farmacología , Trastuzumab/uso terapéutico
8.
Int J Mol Sci ; 23(9)2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35562953

RESUMEN

Isocitrate dehydrogenase (IDH)-wildtype glioblastoma (GBM) is a fast growing and highly heterogeneous tumor, often characterized by the presence of glioblastoma stem cells (GSCs). The plasticity of GSCs results in therapy resistance and impairs anti-tumor immune response by influencing immune cells in the tumor microenvironment (TME). Previously, ß-catenin was associated with stemness in GBM as well as with immune escape mechanisms. Here, we investigated the effect of ß-catenin on attracting monocytes towards GBM cells. In addition, we evaluated whether CCL2 is involved in ß-catenin crosstalk between monocytes and tumor cells. Our analysis revealed that shRNA targeting ß-catenin in GBMs reduces monocytes attraction and impacts CCL2 secretion. The addition of recombinant CCL2 restores peripheral blood mononuclear cells (PBMC) migration towards medium (TCM) conditioned by shß-catenin GBM cells. CCL2 knockdown in GBM cells shows similar effects and reduces monocyte migration to a similar extent as ß-catenin knockdown. When investigating the effect of CCL2 on ß-catenin activity, we found that CCL2 modulates components of the Wnt/ß-catenin pathway and alters the clonogenicity of GBM cells. In addition, the pharmacological ß-catenin inhibitor MSAB reduces active ß-catenin, downregulates the expression of associated genes and alters CCL2 secretion. Taken together, we showed that ß-catenin plays an important role in attracting monocytes towards GBM cells in vitro. We hypothesize that the interactions between ß-catenin and CCL2 contribute to maintenance of GSCs via modulating immune cell interaction and promoting GBM growth and recurrence.


Asunto(s)
Neoplasias Encefálicas , Quimiocina CCL2 , Glioblastoma , beta Catenina , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quimiocina CCL2/genética , Quimiocina CCL2/farmacología , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Monocitos/metabolismo , Microambiente Tumoral , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
9.
J Leukoc Biol ; 112(2): 273-278, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34939227

RESUMEN

Increased levels of the anti-inflammatory peptide Catestatin (CST), a cleavage product of the pro-hormone chromogranin A, correlate with less severe outcomes in hypertension, colitis, and diabetes. However, it is unknown how CST reduces the infiltration of monocytes and macrophages (Mϕs) in inflamed tissues. Here, it is reported that CST blocks leukocyte migration toward inflammatory chemokines. By in vitro and in vivo migration assays, it is shown that although CST itself is chemotactic, it blocks migration of monocytes and neutrophils to inflammatory attracting factor CC-chemokine ligand 2 (CCL2) and C-X-C motif chemokine ligand 2 (CXCL2). Moreover, it directs CX3 CR1+ Mϕs away from pancreatic islets. These findings suggest that the anti-inflammatory actions of CST are partly caused by its regulation of chemotaxis.


Asunto(s)
Quimiotaxis de Leucocito , Quimiotaxis , Antiinflamatorios/farmacología , Quimiocina CCL2/farmacología , Quimiocinas/farmacología , Cromogranina A/farmacología , Ligandos , Neutrófilos , Fragmentos de Péptidos , Péptidos/farmacología
10.
J Tissue Eng Regen Med ; 16(2): 163-176, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34811942

RESUMEN

Human adipose-derived stem cells (hADSCs) and human umbilical vein endothelial cells (HUVECs) co-cultured in vitro are widely used in adipose tissue engineering but exhibit various limitations. Chemokine (C-C motif) ligand 2 (CCL2) has been proved essential during adipogenesis and angiogenesis in vivo. We examined whether adipogenesis and angiogenesis could also be directly promoted by CCL2 in vitro. Cells were cultured with 0, 10, 50, and 100 ng/ml CCL2. The effects of CCL2 on adipogenesis of hADSCs, and lipid accumulation in the positive control group (hADSCs), blank control group (hADSCs + HUVECs), and experimental group (hADSCs + HUVECs + CCL2) in the hADSC and HUVEC direct co-culture system were evaluated by Oil Red O staining. Angiogenesis in the presence of CCL2 was evaluated by Matrigel tube formation assay. Angiogenic- and adipogenic-associated gene and protein expression in the co-culture system were measured by Quantitative Real-time Polymerase Chain Reaction and western blotting, respectively. All concentrations of CCL2 promoted hADSC adipogenic differentiation and HUVEC tube formation (P < 0.05). Following direct co-culture, the experimental group accumulated more lipid droplets than the positive control (P < 0.0001), whereas the latter showed better adipogenesis than the blank control group. 50 ng/ml CCL2 exhibited stronger adipogenic and angiogenic potential than other concentrations. After 72 h of direct co-culture, the mRNA expression of adipogenic differentiation (peroxisome proliferators-activated receptorsγ, CCAAT/enhancer binding protein-α, Leptin, and lipoprotein lipase) and angiogenic genes (vascular endothelial growth factor-A, vascular endothelial growth factor receptor 2, matrix metalloprotein (MMP) 9, and 14) in the experimental group was much higher than in the control (P < 0.05). The addition of 50 ng/ml CCL2 in the system resulted in elevated phosphorylated Protein kinase B/AKT expression. In summary, CCL2 directly promoted adipogenesis of hADSCs and angiogenesis of HUVECs under both mono-culture and co-culture condition in vitro possibly by enhancing AKT phosphorylation. An optimal concentration of 50 ng/ml CCL2 could improve the adipogenesis and angiogenesis of hADSC and HUVEC co-culture system.


Asunto(s)
Quimiocina CCL2 , Factor A de Crecimiento Endotelial Vascular , Adipogénesis , Tejido Adiposo , Células Cultivadas , Quimiocina CCL2/farmacología , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neovascularización Fisiológica , Células Madre , Ingeniería de Tejidos , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Neuroinflammation ; 18(1): 279, 2021 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857006

RESUMEN

BACKGROUND: Opioid analgesics remain widely used for pain treatment despite the related serious side effects. Some of those, such as opioid tolerance and opioid-induced hyperalgesia may be at least partially due to modulation of opioid receptors (OR) function at nociceptive synapses in the spinal cord dorsal horn. It was suggested that increased release of different chemokines under pathological conditions may play a role in this process. The goal of this study was to investigate the crosstalk between the µOR, transient receptor potential vanilloid 1 (TRPV1) receptor and C-C motif ligand 2 (CCL2) chemokine and the involvement of spinal microglia in the modulation of opioid analgesia. METHODS: Patch-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and dorsal root evoked currents (eEPSC) in spinal cord slices superficial dorsal horn neurons were used to evaluate the effect of µOR agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO), CCL2, TRPV1 antagonist SB366791 and minocycline. Paw withdrawal test to thermal stimuli was combined with intrathecal (i.t.) delivery of CCL2 and DAMGO to investigate the modulation in vivo. RESULTS: Application of DAMGO induced a rapid decrease of mEPSC frequency and eEPSC amplitude, followed by a delayed increase of the eESPC amplitude, which was prevented by SB366791. Chemokine CCL2 treatment significantly diminished all the DAMGO-induced changes. Minocycline treatment prevented the CCL2 effects on the DAMGO-induced eEPSC depression, while mEPSC changes were unaffected. In behavioral experiments, i.t. injection of CCL2 completely blocked DAMGO-induced thermal hypoalgesia and intraperitoneal pre-treatment with minocycline prevented the CCL2 effect. CONCLUSIONS: Our results indicate that opioid-induced inhibition of the excitatory synaptic transmission could be severely attenuated by increased CCL2 levels most likely through a microglia activation-dependent mechanism. Delayed potentiation of neurotransmission after µOR activation is dependent on TRPV1 receptors activation. Targeting CCL2 and its receptors and TRPV1 receptors in combination with opioid therapy could significantly improve the analgesic properties of opioids, especially during pathological states.


Asunto(s)
Analgésicos Opioides/farmacología , Quimiocina CCL2/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Nocicepción/efectos de los fármacos , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Anilidas/farmacología , Animales , Cinamatos/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Masculino , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar
12.
Biochem Biophys Res Commun ; 582: 105-110, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34710824

RESUMEN

BACKGROUND: Cell-free heme-containing proteins mediate endothelial injury in a variety of disease states including subarachnoid hemorrhage and sepsis by increasing endothelial permeability. Inflammatory cells are also attracted to sites of vascular injury by monocyte chemotactic protein 1 (MCP-1) and other chemokines. We have identified a novel peptide hormone, adropin, that protects against hemoglobin-induced endothelial permeability and MCP-1-induced macrophage migration. METHODS: Human microvascular endothelial cells were exposed to cell-free hemoglobin (CFH) with and without adropin treatment before measuring monolayer permeability using a FITC-dextran tracer assay. mRNA and culture media were collected for molecular studies. We also assessed the effect of adropin on macrophage movement across the endothelial monolayer using an MCP-1-induced migration assay. RESULTS: CFH exposure decreases adropin expression and increases paracellular permeability of human endothelial cells. Treating cells with synthetic adropin protects against the increased permeability observed during the natural injury progression. Cell viability was similar in all groups and Hmox1 expression was not affected by adropin treatment. MCP-1 potently induced macrophage migration across the endothelial monolayer and adropin treatment effectively reduced this phenomenon. CONCLUSIONS: Endothelial injury is a hallmark of many disease states. Our results suggest that adropin treatment could be a valuable strategy in preventing heme-mediated endothelial injury and macrophage infiltration. Further investigation of adropin therapy in animal models and human tissue specimens is needed.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/antagonistas & inhibidores , Células Endoteliales/efectos de los fármacos , Hemoglobinas/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intercelular/farmacología , Macrófagos/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Quimiocina CCL2/farmacología , Citoprotección/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Hemoglobinas/farmacología , Humanos , Macrófagos/citología
13.
Cytokine ; 142: 155503, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33781652

RESUMEN

The role of glycosaminoglycans on the surface of immune cells has so far been less studied compared to their participation in inflammatory responses as members of the endothelium and the extracellular matrix. In this study we have therefore investigated if glycosaminoglycans on immune cells act in concert with GPC receptors (i.e. both being cis-located on leukocytes) in chemokine-induced leukocyte mobilisation. For this purpose, freshly-prepared human neutrophils and monocytes were treated with heparinase III or chondroitinase ABC to digest heparan sulfate -chains or chondroitin sulfate-chains, respectively, from the leukocyte surfaces. Subsequent analysis of CXCL8- and CCL2-induced chemotaxis revealed that leukocyte migration was strongly reduced after eliminating heparan sulfate from the surface of neutrophils and monocytes. In the case of monocytes, an additional dependence of CCL2-induced chemotaxis on chondroitin sulfate was observed. We compared these results with the effect on chemotaxis of a heparan sulfate masking antibody and obtained similarly reduced migration. Following our findings, we postulate that glycosaminoglycans located on target leukocytes act synergistically with GPC receptors on immune cell migration, which is further influenced by glycosaminoglycans located on the inflamed tissue (i.e. trans with respect to the immune cell/GPC receptor). Both glycosaminoglycan localization sites seem to be important during inflammatory processes and could potentially be tackled in chemokine-related diseases.


Asunto(s)
Movimiento Celular , Quimiocina CCL2/farmacología , Glicosaminoglicanos/metabolismo , Interleucina-8/farmacología , Monocitos/metabolismo , Neutrófilos/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Condroitinasas y Condroitín Liasas/metabolismo , Femenino , Glipicanos/genética , Glipicanos/metabolismo , Liasa de Heparina/metabolismo , Humanos , Monocitos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos , Sindecanos/genética , Sindecanos/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos
14.
mBio ; 12(2)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33727362

RESUMEN

The human immunodeficiency virus (HIV) enters the central nervous system (CNS) within a few days after primary infection, establishing viral reservoirs that persist even with combined antiretroviral therapy (cART). We show that monocytes from people living with HIV (PLWH) on suppressive cART harboring integrated HIV, viral mRNA, and/or viral proteins preferentially transmigrate across the blood-brain barrier (BBB) to CCL2 and are significantly enriched post-transmigration, and even more highly enriched posttransmigration than T cells with similar properties. Using HIV-infected ART-treated mature monocytes cultured in vitro, we recapitulate these findings and demonstrate that HIV+ CD14+ CD16+ ART-treated monocytes also preferentially transmigrate. Cenicriviroc and anti-JAM-A and anti-ALCAM antibodies significantly and preferentially reduce/block transmigration of HIV+ CD14+ CD16+ ART-treated monocytes. These findings highlight the importance of monocytes in CNS HIV reservoirs and suggest targets to eliminate their formation and reseeding.IMPORTANCE We characterized mechanisms of CNS viral reservoir establishment/replenishment using peripheral blood mononuclear cells (PBMC) of PLWH on cART and propose therapeutic targets to reduce/block selective entry of cells harboring HIV (HIV+) into the CNS. Using DNA/RNAscope, we show that CD14+ CD16+ monocytes with integrated HIV, transcriptionally active, and/or with active viral replication from PBMC of PLWH prescribed cART and virally suppressed, selectively transmigrate across a human BBB model. This is the first study to our knowledge demonstrating that monocytes from PLWH with HIV disease for approximately 22 years and with long-term documented suppression can still carry virus into the CNS that has potential to be reactivated and infectious. This selective entry into the CNS-and likely other tissues-indicates a mechanism of reservoir formation/reseeding in the cART era. Using blocking studies, we propose CCR2, JAM-A, and ALCAM as targets on HIV+ CD14+ CD16+ monocytes to reduce and/or prevent CNS reservoir replenishment and to treat HAND and other HIV-associated comorbidities.


Asunto(s)
Sistema Nervioso Central/virología , Reservorios de Enfermedades/virología , Leucocitos Mononucleares/fisiología , Leucocitos Mononucleares/virología , Migración Transendotelial y Transepitelial/inmunología , Antirretrovirales/farmacología , Antirretrovirales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Asparaginasa/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/virología , Ensayos de Migración de Leucocitos , Sistema Nervioso Central/efectos de los fármacos , Quimiocina CCL2/inmunología , Quimiocina CCL2/farmacología , Citarabina/uso terapéutico , Daunorrubicina/uso terapéutico , Femenino , Infecciones por VIH/virología , Humanos , Técnicas In Vitro , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Persona de Mediana Edad , Tioguanina/uso terapéutico
15.
BMC Dev Biol ; 20(1): 20, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33023470

RESUMEN

BACKGROUND: Monocyte chemoattractant protein-1(MCP-1) is a chemokine secreted by Leydig cells and peritubular myoid cells in the rat testis. Its role in regulating the development of Leydig cells via autocrine and paracrine is still unclear. The objective of the current study was to investigate the effects of MCP-1 on Leydig cell regeneration from stem cells in vivo and on Leydig cell development in vitro. RESULTS: Intratesticular injection of MCP-1(10 ng/testis) into Leydig cell-depleted rat testis from post-EDS day 14 to 28 significantly increased serum testosterone and luteinizing hormone levels, up-regulated the expression of Leydig cell proteins, LHCGR, SCARB1, CYP11A1, HSD3B1, CYP17A1, and HSD17B3 without affecting progenitor Leydig cell proliferation, as well as increased ERK1/2 phosphorylation. MCP-1 (100 ng/ml) significantly increased medium testosterone levels and up-regulated LHCGR, CYP11A1, and HSD3B1 expression without affecting EdU incorporation into stem cells after in vitro culture for 7 days. RS102895, a CCR2 inhibitor, reversed MCP-1-mediated increase of testosterone level after culture in combination with MCP-1. CONCLUSION: MCP-1 stimulates the differentiation of stem and progenitor Leydig cells without affecting their proliferation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quimiocina CCL2/farmacología , Células Intersticiales del Testículo/citología , Regeneración/efectos de los fármacos , Células Madre/citología , Testículo/fisiología , Animales , Expresión Génica/efectos de los fármacos , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/sangre , Masculino , Ratas , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Testículo/efectos de los fármacos , Testosterona/sangre
16.
Front Immunol ; 11: 2129, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072075

RESUMEN

Macrophages are key targets of human immunodeficiency virus type 1 (HIV-1) infection and main producers of the proinflammatory chemokine CC chemokine ligand 2 (CCL2), whose expression is induced by HIV-1 both in vitro and in vivo. We previously found that CCL2 neutralization in monocyte-derived macrophages (MDMs) strongly inhibited HIV-1 replication affecting post-entry steps of the viral life cycle. Here, we used RNA-sequencing to deeply characterize the cellular factors and pathways modulated by CCL2 blocking in MDMs and involved in HIV-1 replication restriction. We report that exposure to CCL2 neutralizing antibody profoundly affected the MDM transcriptome. Functional annotation clustering of up-regulated genes identified two clusters enriched for antiviral defense and immune response pathways, comprising several interferon-stimulated, and restriction factor coding genes. Transcripts in the clusters were enriched for RELA and NFKB1 targets, suggesting the activation of the canonical nuclear factor κB pathway as part of a regulatory network involving miR-155 up-regulation. Furthermore, while HIV-1 infection caused small changes to the MDM transcriptome, with no evidence of host defense gene expression and type I interferon signature, CCL2 blocking enabled the activation of a strong host innate response in infected macrophage cultures, and potently inhibited viral genes expression. Notably, an inverse correlation was found between levels of viral transcripts and of the restriction factors APOBEC3A (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3 A), ISG15, and MX1. These findings highlight an association between activation of innate immune pathways and HIV-1 restriction upon CCL2 blocking and identify this chemokine as an endogenous factor contributing to the defective macrophage response to HIV-1. Therapeutic targeting of CCL2 may thus strengthen host innate immunity and restrict HIV-1 replication.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Quimiocina CCL2/farmacología , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica/efectos de los fármacos , VIH-1/genética , Inmunidad Innata , Macrófagos/metabolismo , Anticuerpos Neutralizantes/inmunología , Especificidad de Anticuerpos , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/inmunología , Citidina Desaminasa/fisiología , Conjuntos de Datos como Asunto , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , MicroARNs/biosíntesis , MicroARNs/genética , Anotación de Secuencia Molecular , FN-kappa B/metabolismo , Proteínas/fisiología , ARN Viral/biosíntesis , ARN Viral/genética , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Latencia del Virus , Replicación Viral
17.
J Pharmacol Sci ; 144(2): 61-68, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32684333

RESUMEN

The effects of adipokine administration to the hypothalamic preoptic area (POA), which is one of the body temperature (BT) regulation centers in the central nervous system, on BT were investigated in male Wistar rats. BT was measured in conscious rats using telemetry. Insulin-like growth factor-1 (IGF-1), interleukin-1ß (IL-1ß), monocyte chemoattractant protein-1 and lipocalin-2 produced hyperthermia, and the effects induced by IL-1ß (25 ng) and IGF-1 (5 µg) were sustainable and remarkable. IL-6 did not show any significant effect. The IGF-1-induced effect was inhibited by pretreatment with IGF binding protein 3 (IGFBP3) or NVP-AEW541 (NVP, a selective inhibitor of type 1 IGF receptor tyrosine kinase, IGF1R TK). NVP-induced inhibition was observed only in the early phase of IGF-1-induced hyperthermia. In addition, IGF-1 increased the IL-1ß concentration in the microdialysate of POA perfusion, but did not increase the IL-1ß concentration in the plasma or the PGE2 concentration in the microdialysate. These findings suggested that IGF-1 produced hyperthermia, which was mediated, at least a part, through an increased IL-1ß concentration after activation of IGF1R TK in the POA, and the IGF-IGFBP system possibly participates in BT homeostasis in the POA.


Asunto(s)
Adipoquinas/administración & dosificación , Adipoquinas/farmacología , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/genética , Área Preóptica/metabolismo , Área Preóptica/fisiología , Animales , Quimiocina CCL2/administración & dosificación , Quimiocina CCL2/farmacología , Fiebre/inducido químicamente , Fiebre/genética , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/farmacología , Interleucina-1beta/administración & dosificación , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Lipocalina 2/administración & dosificación , Lipocalina 2/farmacología , Masculino , Proteínas Tirosina Quinasas/metabolismo , Ratas Wistar , Receptor IGF Tipo 1/metabolismo
18.
Mol Med Rep ; 22(1): 97-104, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32377737

RESUMEN

Excessive numbers of osteoclasts are responsible for inflammation­induced osteolysis. Identification of osteoclast­targeting agents may facilitate the development of a novel therapeutic approach for the treatment of pathological bone loss. Seven­amino acid truncated (7ND) protein, a mutant form of monocyte chemoattractant protein­1 (MCP­1), functions as a competitive inhibitor of MCP­1. However, the effects of 7ND protein on osteoclast differentiation remain unknown. Therefore, in the present study, the effects of 7ND protein on osteoclast differentiation induced by tumour necrosis factor superfamily member 11 were investigated. In the present study, 7ND protein inhibited the osteoclast differentiation of peripheral blood mononuclear cells without influencing cell proliferation. Furthermore, to evaluate the effects of 7ND protein in vivo, a lipopolysaccharide (LPS)­induced calvarial bone erosion animal model was established. The 7ND protein remarkably attenuated LPS­induced bone resorption, as assessed by micro­computed tomography and histological analysis. Taken together, the present results suggested the feasibility of local delivery of 7ND protein to mitigate osteoclast differentiation and LPS­induced osteolysis, which may represent a potential approach to treat inflammatory bone destruction.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Quimiocina CCL2/uso terapéutico , Osteogénesis/efectos de los fármacos , Osteólisis/tratamiento farmacológico , Adulto , Animales , Resorción Ósea/etiología , Resorción Ósea/patología , Células Cultivadas , Quimiocina CCL2/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Lipopolisacáridos/efectos adversos , Masculino , Ratones , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteólisis/etiología , Osteólisis/patología , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
19.
PLoS One ; 15(3): e0230080, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32155215

RESUMEN

Human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) have shown potential in facilitating recovery from spinal cord injury (SCI) through communicating with microglia/macrophages (MG/MΦ). We here focused on chemokines as a candidate for the communication. Selected MG/MΦ-related chemokines were determined gene expression after SCI and further focused CCL2/CCR2 and CCL5/CCR5 to estimate role of the chemokines by hMSCs. Male C57/BL6 mice were subjected to spinal cord transection. Gene expression was assayed in the spinal cords following SCI for selected MG/MΦ-related chemokines and their receptors. hMSCs (5×105 cells) were then transplanted into parenchyma of the spinal cord, and the expressions of the Ccl2/Ccr2 and Ccl5/Ccr5 axes, inflammation, MG/MΦ-polarization, and axonal regeneration were evaluated to measure the influence of the hMSCs. Finally, mouse CCL5 was injected into the spinal cords. Acute increases in gene expression after SCI were observed for most chemokines, including Ccl2; chronic increases were observed for Ccl5. CCL2+-cells merged with NeuN+-neurons. CCR2+ immunoreactivity was principally observed in Ly-6G+/iNOS+-granulocytes on postoperative day (pod) 1, and CCL5+ and CCR5+ immunoreactivity overlapped with NeuN+-neurons and F4/80+-MG/MΦ on pod 14. The hMSC transplantation enhanced Ccl2 and Ccl5 and improved locomotor activity. The hMSC implantation did not alter the number of Ly-6G+/CCR2+ but decreased Il1, Elane, and Mpo on pod 3. Conversely, hMSC transplantation increased expression of Zc3h12a (encodes MCP-1-induced protein) on pod 14. Moreover, hMSC increased the Aif1, and two alternatively activated macrophage (AAM)-related genes, Arg1 and Chil3 (Ym1), as well as axonal regenerative markers, Dpysl2 and Gap43. Gene expression indicative of AAM polarization and axonal regeneration were partially recovered by CCL5 injection. These results suggest that hMSC implantation increases Ccl2 and Ccl5, improves locomotor activity, enhances MG/MΦ polarization to AAM, and increases the gene expression of axonal regenerative markers. These functions of hMSCs might be partially mediated by the CCL2/CCR2 and CCL5/CCR5 axes.


Asunto(s)
Axones/patología , Quimiocina CCL2/farmacología , Quimiocina CCL5/farmacología , Trasplante de Células Madre Mesenquimatosas , Traumatismos de la Médula Espinal/terapia , Animales , Axones/efectos de los fármacos , Quimiocina CCL2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Receptores CCR2/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
20.
Front Immunol ; 10: 1200, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191554

RESUMEN

Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS), characterized by the infiltration of mononuclear cells into the CNS and a subsequent inflammation of the brain. Monocytes are implicated in disease pathogenesis not only in their function as potential antigen-presenting cells involved in the local reactivation of encephalitogenic T cells but also by independent effector functions contributing to structural damage and disease progression. However, monocytes also have beneficial effects as they can exert anti-inflammatory activity and promote tissue repair. Glucocorticoids (GCs) are widely used to treat acute relapses in MS patients. They act on a variety of cell types but their exact mechanisms of action including their modulation of monocyte function are not fully understood. Here we investigated effects of the therapeutically relevant GC methylprednisolone (MP) on monocytes from healthy individuals and MS patients in vitro and in vivo. The monocyte composition in the blood was different in MS patients compared to healthy individuals, but it was only marginally affected by MP treatment. In contrast, application of MP caused a marked shift toward an anti-inflammatory monocyte phenotype in vitro and in vivo as revealed by an altered gene expression profile. Chemotaxis of monocytes toward CCL2, CCL5, and CX3CL1 was increased in MS patients compared to healthy individuals and further enhanced by MP pulse therapy. Both of these migration-promoting effects were more pronounced in MS patients with an acute relapse than in those with a progressive disease. Interestingly, the pro-migratory GC effect was independent of chemokine receptor levels as exemplified by results obtained for CCR2. Collectively, our findings suggest that GCs polarize monocytes toward an anti-inflammatory phenotype and enhance their migration into the inflamed CNS, endowing them with the capacity to suppress the pathogenic immune response.


Asunto(s)
Antiinflamatorios/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Metilprednisolona/farmacología , Monocitos/efectos de los fármacos , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Adulto , Anciano , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/genética , Quimiocina CCL2/farmacología , Quimiocina CCL2/fisiología , Femenino , Humanos , Masculino , Metilprednisolona/administración & dosificación , Metilprednisolona/uso terapéutico , Persona de Mediana Edad , Monocitos/inmunología , Esclerosis Múltiple Crónica Progresiva/inmunología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Quimioterapia por Pulso , Receptores CCR2/biosíntesis , Receptores CCR2/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Receptores de Glucocorticoides/biosíntesis , Receptores de Glucocorticoides/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...