Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cells ; 13(5)2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38474420

ABSTRACT

NAD+ boosting via nicotinamide riboside (NR) confers anti-inflammatory effects. However, its underlying mechanisms and therapeutic potential remain incompletely defined. Here, we showed that NR increased the expression of CC-chemokine receptor 7 (CCR7) in human M1 macrophages by flow cytometric analysis of cell surface receptors. Consequently, chemokine ligand 19 (CCL19, ligand for CCR7)-induced macrophage migration was enhanced following NR administration. Metabolomics analysis revealed that prostaglandin E2 (PGE2) was increased by NR in human monocytes and in human serum following in vivo NR supplementation. Furthermore, NR-mediated upregulation of macrophage migration through CCL19/CCR7 was dependent on PGE2 synthesis. We also demonstrated that NR upregulated PGE2 synthesis through SIRT3-dependent post-transcriptional regulation of cyclooxygenase 2 (COX-2). The NR/SIRT3/migration axis was further validated using the scratch-test model where NR and SIRT3 promoted more robust migration across a uniformly disrupted macrophage monolayer. Thus, NR-mediated metabolic regulation of macrophage migration and wound healing may have therapeutic potential for the topical management of chronic wound healing.


Subject(s)
Dinoprostone , Niacinamide/analogs & derivatives , Pyridinium Compounds , Sirtuin 3 , Humans , Dinoprostone/metabolism , Ligands , Receptors, CCR7/metabolism , Macrophages/metabolism
2.
Altern Ther Health Med ; 29(3): 160-165, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36735714

ABSTRACT

Context: Gastric cancer (GC) remains one of the most prevalent malignancies worldwide, and no effective cure exists for advanced GC. Clinicians believe that molecularly targeted therapy through PCGs may replace surgery, radiotherapy, and other treatments as a breakthrough in curing malignancies. Objective: The study intended to examine the impact of aberrant expression of the protein-coding genes (PCGs) associated with regulatory T cells on the prognosis of patients with gastric cancer (GC). Design: The research team performed a genetic study through research of genetic data in online databases. Setting: The study took place at Zhongda Hospital. Outcome Measures: The research team selected a publicly available dataset, genetic suppressor element 109476 (GSE109476), from the Gene Expression Omnibus (GEO) database for differential gene analysis, gene ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to screen for PCGs associated with regulatory T cells as well as the Gene Expression Profiling Interactive Analysis (GEPIA) database with the Kaplan-Meier Plotter database to analyze the expression of the above PCGs in GC and the prognostic impact on GC. Results: The GEO2R analysis found 315 differentially expressed PCGs in GSE109476, among which nine PCGs were associated with regulatory T cells: (1) chemokine (C-C motif) ligand 19 (CCL19), (2) CCL21, (3) C-C chemokine receptor type 7 (CCR7), (4) cluster of differentiation 70 (CD70), (5) ephrin B3 (EFNB3), (6) early growth response 3 (EGR3), (7) interleukin-7 receptor (IL7R), (8) galectin-1 (LGALS1), and (9) tumor necrosis factor (TNF) receptor superfamily member 13C (TNFRSF13C). The GEPIA database indicated that no significant differences existed between the expression of CCL19, CCL21, CD70, EFNB3, EGR3, IL7R, and TNFRSF13C in stomach adenocarcinoma (STAD) tissues and that in normal tissues (P > .05), while expressions of CCR7 and LGALS1 were significantly elevated in STAD tissues compared to the normal tissues (P < .05). The Kaplan-Meier Plotter database analysis, on the other hand, showed a significant relationship between all of the above-mentioned PCGs, except CCL19, and the prognosis of GC. Conclusions: CCL19, CCL21, CCR7, CD70, EFNB3, EGR3, IL7R, LGALS1, and TNFRSF13C are PCGs are differentially expressed in GC and closely associated with regulatory T cells. They may affect the occurrence and development of GC through a variety of pathways, including regulation of immune infiltration and inflammation, and are of great potential research value.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Galectin 1 , Receptors, CCR7 , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Ephrin-B3
3.
J Cosmet Dermatol ; 22(5): 1602-1612, 2023 May.
Article in English | MEDLINE | ID: mdl-36639978

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a chronic inflammatory skin disease with a genetic predisposition, and the traditional Chinese medicine Morinda officinalis and its roots are characterized with anti-inflammatory effects and have been used for the treatment of various disease. However, it is still largely unknown whether Morinda officinalis extract (MOE) can be used for the treatment of AD. OBJECTIVES: In our study we aimed to determine whether MOE could ameliorate 2,4-dinitrochlorobenzene (DNCB)-induced AD and elucidate molecular mechanisms. METHODS: We established an AD mouse model by using DNCB. Skin pathological analysis and ELISA assay were used to detect the effect of MOE on the inflammation of AD model mouse skin and the expression changes of inflammatory factors, and further functional verification was performed in TNF-α/IFN-γ-induced HaCaT cells. RESULTS: Our in vivo experiments confirmed that MOE remarkably reduced DNCB-induced AD lesions and symptoms, such as epidermal and dermal thickness and mast cell infiltration and inflammatory cytokines secretion in the mice models. In addition, the underlying mechanisms by which MOE ameliorated AD had been uncovered, and we verified that MOE inhibited MALAT1 expression in AD, resulting in attenuated expression of C-C chemokine receptor type 7 (CCR7) regulated by MALAT1-sponge miR-590-5p in a competing endogenous RNA (ceRNA) mechanisms-dependent manner, thereby inhibiting TNF-α/IFN-γ-induced cellular proliferation and inflammation.


Subject(s)
Dermatitis, Atopic , MicroRNAs , Morinda , RNA, Long Noncoding , Animals , Mice , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Morinda/metabolism , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha/metabolism , Dinitrochlorobenzene/metabolism , Dinitrochlorobenzene/pharmacology , Dinitrochlorobenzene/therapeutic use , Receptors, CCR7/metabolism , Receptors, CCR7/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Skin/metabolism , Inflammation/pathology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Extracts/metabolism , Cytokines/metabolism
4.
Cancer Immunol Immunother ; 71(3): 627-636, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34297159

ABSTRACT

CAP-100 is a novel therapeutic antibody directed against the ligand binding site of human CCR7. This chemokine receptor is overexpressed in chronic lymphocytic leukemia (CLL) and orchestrates the homing of CLL cells into the lymph node. Previous studies, on a very limited number of samples, hypothesized that the Bruton's tyrosine kinase inhibitor (BTKi) ibrutinib might induce loss of surface CCR7 levels in CLL cells. CAP-100 will be evaluated in clinical trials as a therapy for relapse/refractory CLL patients, who have received at least two systemic therapies (NCT04704323). As nowadays many relapse/refractory CLL patients will have received ibrutinib as a prior therapy, we aimed to investigate in a large cohort of CLL patients the impact of this BTKi on CCR7 expression and functionality as well as on the therapeutic activity of CAP-100. Our data confirm that ibrutinib moderately down-regulates the very high expression of CCR7 in CLL cells but has no apparent effect on CCR7-induced chemotaxis. Moreover, CLL cells are perfectly targetable by CAP-100 which led to a complete inhibition of CCR7-mediated migration and induced strong target cell killing through antibody-dependent cell-mediated cytotoxicity, irrespective of previous or contemporary ibrutinib administration. Together, these results validate the therapeutic utility of CAP-100 as a next-line single-agent therapy for CLL patients who failed to ibrutinib and confirm that CAP-100 and ibrutinib have complementary non-overlapping mechanisms of action, potentially allowing for combination therapy.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents, Immunological/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptors, CCR7/genetics , Adenine/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, CCR7/antagonists & inhibitors , Receptors, CCR7/metabolism
5.
Molecules ; 26(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34770763

ABSTRACT

Homeostatic trafficking of immune cells by CC chemokine receptor 7 (CCR7) keeps immune responses and tolerance in a balance. The involvement of this protein in lymph node metastasis in cancer marks CCR7 as a penitential drug target. Using the crystal structure of CCR7, herein, a comprehensive virtual screening study is presented to filter novel strong CCR7 binding phytochemicals from Saudi medicinal plants that have a higher binding affinity for the intracellular allosteric binding pocket. By doing so, three small natural molecules named as Hit-1 (1,8,10-trihydroxy-3-methoxy-6-methylanthracen-9(4H)-one), Hit-2 (4-(3,4-dimethoxybenzyl)-3-(4-hydroxy-3-methoxybenzyl)dihydrofuran-2(3H)-one), and Hit-3 (10-methyl-12,13-dihydro-[1,2]dioxolo[3,4,5-de]furo[3,2-g]isochromeno[4,3-b]chromen-8-ol) are predicted showing strong binding potential for the CC chemokine receptor 7 allosteric pocket. During molecular dynamics simulations, the compounds were observed in the formation of several chemical bonding of short bond distances. Additionally, the molecules remained in strong contact with the active pocket residues and experienced small conformation changes that seemed to be mediated by the CCR7 loops to properly engage the ligands. Two types of binding energy methods (MM/GBPBSA and WaterSwap) were additionally applied to further validate docking and simulation findings. Both analyses complement the good affinity of compounds for CCR7, the electrostatic and van der Waals energies being the most dominant in intermolecular interactions. The active pocket residue's role in compounds binding was further evaluated via alanine scanning, which highlighted their importance in natural compounds binding. Additionally, the compounds fulfilled all drug-like rules: Lipinski, Ghose, Veber, Egan, and Muegge passed many safety parameters, making them excellent anti-cancer candidates for experimental testing.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Phytochemicals/chemistry , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Receptors, CCR7/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Drug Discovery , Humans , Molecular Conformation , Molecular Structure , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Protein Binding , Receptors, CCR7/antagonists & inhibitors , Saudi Arabia
6.
Biosci Biotechnol Biochem ; 84(9): 1861-1869, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32475338

ABSTRACT

Dendritic cells (DCs) are play critical roles in the priming and regulation of immune responses. DCs rapidly process and convey these antigens to prime antigen-specific T cells. Therefore, regulation of DCs functions is important for immunity and immunotherapies. Immune adjuvants for DCs activation are needed to improve the efficacy of vaccines against tumors and many infectious diseases. Therefore, we demonstrate that H. fusiformis extract can regulate DCs maturation and activation. H. fusiformis extract induced costimulatory molecules (CD 80 and CD86), antigen-presenting molecules (major histocompatibility complex (MHC) I and II), CCR7 expression, and interleukin (IL)-12 production in DCs. These effects are associated with upregulation of mitogen-activated protein kinase (MAPK) signaling pathway. In addition, H. fusiformis extract induces costimulatory molecules on splenic DCs and activated CD8+ T cells in vivo. Taken together, these findings suggest that H. fusiformis extract may be a potential efficient immune therapeutic compound in DCs-mediated immunotherapies. ABBREVIATIONS: CTL: cytotoxic T lymphocytes; DCs: dendritic cells; ERK: extracellular signal-regulated kinases; IL: interleukini; JNK: c-Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; MHC: major histocompatibility complex.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/drug effects , Plant Extracts/pharmacology , Sargassum/chemistry , Cell Differentiation/drug effects , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-12/biosynthesis , Lymphocyte Activation/drug effects , MAP Kinase Signaling System/drug effects , Receptors, CCR7/metabolism
7.
Phytomedicine ; 64: 153084, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31514083

ABSTRACT

BACKGROUND: Metastasized melanoma is extremely difficult to treat. Activation of C-C chemokine receptor type 7 (CCR7) has been linked to melanoma metastasis. CCR7 can be directly regulated by miR-let-7. We have previously shown that an ethanolic extract of an herbal formula comprising Sophorae Flos and Lonicerae Japonicae Flos (SLE) inhibits melanoma cell migration and invasion. PURPOSE: In this study, we determined whether SLE suppresses melanoma metastasis, and whether regulation of miR-let-7a/f-CCR7 signaling is involved in the effect. STUDY DESIGN AND METHODS: Small RNA sequencing was conducted to compare miRNA expression profiles of B16F10 tumors dissected from SLE-treated or untreated mice. Western blot and RT-qPCR analyses were employed to examine protein and miRNA levels, respectively. A B16F10 melanoma lung metastasis mouse model was used to evaluate the effects of SLE on melanoma metastasis. MiR-let-7a/f-knockdown and CCR7-overexpression cell models were used to investigate the involvement of miR-let-7a/f-CCR7 signaling in the anti-metastatic effects of SLE. RESULTS: It was found that SLE upregulated levels of miR-let-7a/f in B16F10 melanoma tissues. SLE significantly elevated levels of miR-let-7a/f, lowered the protein level of CCR7, inhibited the phosphorylation of CCR7 downstream molecules p38 and JNK in B16F10 and A375 melanoma cells. SLE inhibited B16F10 melanoma lung metastasis in mice. SLE upregulated levels of miR-let-7a/f, and lowered protein levels of CCR7, MMP-2, MMP-9, phospho-p38 (Thr180/Tyr182) and phospho-JNK (Thr183/Tyr185) in melanoma-invaded lung tissues. Knockdown of miR-let-7a/f diminished the effects of SLE on CCR7 signaling in, and invasion of, melanoma cells. Overexpression of CCR7 lessened the effects of SLE in inhibiting the phosphorylation of p38 and JNK in, and the invasive capability of, melanoma cells. CONCLUSION: We for the first time demonstrated that SLE inhibits melanoma metastasis in mice, and that regulation of the miR-let-7a/f-CCR7 pathway contributes to the anti-metastatic mechanisms of SLE. These findings provide a pharmacological basis for developing SLE as a modern agent for treating metastatic melanoma. Additionally and importantly, this study suggests that regulating the miR-let-7a/f-CCR7 pathway is a novel strategy for controlling melanoma metastasis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/pharmacology , Melanoma, Experimental/drug therapy , MicroRNAs/metabolism , Plant Extracts/pharmacology , Receptors, CCR7/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Drugs, Chinese Herbal/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lonicera , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Plant Extracts/chemistry , Receptors, CCR7/genetics , Sophora/chemistry
8.
Nutrients ; 11(2)2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30781353

ABSTRACT

Curcumae radix is the dry root of Curcuma longa L. (turmeric) that can be used either as a spice or traditional medicine. The aim of this study was to investigate the survival benefits and the anti-metastatic activity of curcumae radix extract (CRE) in MCF7 cells and in MMTV-PyMT transgenic mice-a mouse model of breast cancer metastasis. In vitro wound scratch assay revealed that CRE treatment inhibited cell motility and cell migration in a dose-dependent manner. To investigate the effect of CRE in breast cancer metastasis, MMTV-PyMT transgenic female virgin mice were used and randomly divided into two groups. For survival curve analysis, CRE was administered in a dose of 50 mg/kg to 8⁻20-week-old mice. Interestingly, CRE treatment significantly increased the median and prolonged survival of MMTV-PyMT mice. Furthermore, CRE treatment decreased tumor burden and inhibited cell proliferation in primary breast tumor, and also suppressed mammary tumor-derived lung metastasis. The size of the lung metastases substantially decreased in the CRE-treated group compared with the ones in the control group. Curcumae radix extract showed anti-metastatic activity through regulating the expression of metastasis markers including C-C Chemokine Receptor Type 7, Matrix Metalloproteinase 9 and the proto-oncogenes c-fos and c-jun. We demonstrated that these metastatic regulators were decreased when CCR7 expression was suppressed in MCF7 cells transfected with CCR7 siRNA. The results of this study show that curcumae radix exerts antitumor and anti-metastatic activities, and we suggest that curcumae radix might be a potential supplement for the treatment and prevention of breast cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Curcuma , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/drug therapy , Neoplasm Metastasis/prevention & control , Plant Extracts/pharmacology , Receptors, CCR7/drug effects , Animals , Female , Genes, fos/drug effects , Genes, jun/drug effects , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/drug effects , Mice , Mice, Transgenic , Plant Roots , Receptors, CCR7/biosynthesis
9.
Interdiscip Sci ; 10(2): 346-366, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28271460

ABSTRACT

The Human Chemokine (C-C motif) ligand 19 (CCL19) protein plays a major role in rheumatic and autoimmune diseases. The 3D models of the CCL19 and its receptor CCR7 are generated using homology modeling and are validated using standard computational protocols. Disulfide bridges identified in 3D model of CCL19 protein give extra stability to the overall protein structure. The active site region of protein CCL19, containing N-terminal amino acid residues (Gly22 to Leu31), is predicted using in silico techniques. Protein-protein docking studies are carried out between the CCL19 and CCR7 proteins to analyse the active site binding interactions of CCL19. The binding domain of CCL19 is subjected to structure-based virtual screening of small molecule databases, and identified several bioisosteric ligand molecules having pyrrolidone and piperidone pharmacophores. The prioritized ligands with acceptable ADME properties are reported as new leads for the design of potential CCL19 antagonists for rheumatic and autoimmune disease therapies.


Subject(s)
Autoimmune Diseases/drug therapy , Chemokine CCL19/chemistry , Chemokine CCL19/metabolism , Computer Simulation , Receptors, CCR7/chemistry , Receptors, CCR7/metabolism , Rheumatic Diseases/drug therapy , Amino Acid Sequence , Catalytic Domain , Conserved Sequence , Disulfides/metabolism , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Protein Domains , Protein Structure, Secondary , Solvents , Structural Homology, Protein
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(10): 1357-1363, 2016 Oct 20.
Article in Chinese | MEDLINE | ID: mdl-27777198

ABSTRACT

OBJECTIVE: To investigate the effect of low-selenium diet on the liver and kidneys of rats and explore the role of macrophage polarization into M1 and M2 phenotypes in liver and kidney injuries. METHODS: Twenty-four rats (12 female and 12 male) were randomly divided into control group and low-selenium group and fed with normal chow (dietary selenium of 0.18 mg/kg) and low-selenium diet (dietary selenium of 0.02 mg/kg) for 109 days. After the feeding, the rats were sacrificed for HE staining to observe liver and kidney pathologies, and immunohistochemistry was performed for analyzing CCR7, CD206, CD163-positive cell numbers in the liver and kidneys. RESULTS: The rats in low-selenium group showed severer fibrosis in the liver and kidney than the control group. In either male or female rats in low-selenium group, CCR7 and CD206 expressions in the liver were comparable with those in control group, but CD163 expression was lower than that in the control group (P<0.05 for both female and male rats). In the kidney, the proximal tubule showed a slightly higher while the distal tubule showed a slightly lower CCR7 expression in low selenium group than in the control group (P>0.05). In low-selenium group, a significantly lower CD163 expression in the distal tubule and a significantly higher CD206 expression in the proximal tubule were noted as compared with the control group (P<0.05 in both female and male rats). Compared with the control rats, the male rats in low-selenium group, but not the female rats, showed a significantly lower CD163 expression in the proximal tubule of the kidney (P<0.05); the female but not the male rats in low-selenium group show a higher CD206 expression in the distal tubule (P<0.05). CONCLUSION: Low-selenium diet can cause liver and kidney fibrosis in rats and may inhibit macrophage activation into the M2 phenotype.


Subject(s)
Diet , Kidney/metabolism , Liver/metabolism , Macrophage Activation , Selenium/administration & dosage , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Female , Fibrosis , Kidney/pathology , Lectins, C-Type/metabolism , Liver/pathology , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Rats , Receptors, CCR7/metabolism , Receptors, Cell Surface/metabolism
11.
Int J Mol Med ; 38(5): 1541-1548, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666000

ABSTRACT

Baicalin, extracted and purified from the Chinese medicinal plant, Scutellaria baicalensis Georgi (Huang qin in Chinese), exhibits potent anti-inflammatory activity against asthma. However, it remains unknown whether baicalin inhibits the activity of CC chemokine receptor 7 (CCR7) and its ligands, which are crucial for the initiation of airway inflammation. In the present study, we investigated the effects of baicalin on CCR7 and its ligands, CCL19 and CCL21, as well as on the nuclear factor-κB (NF-κB) pathway in a mouse model of asthma. A mouse model of acute asthma was established by exposing the mice to ovalbumin (OVA) (by intraperitoneal injection and inhalational challenge). Within 24 h of the final OVA challenge, lung function was detected by direct airway resistance analysis. Lung tissues were examined for pathological changes. Inflammatory cell counts in bronchoalveolar lavage fluid (BALF) were assessed. ELISA was utilized to evaluate the OVA-IgE, CCL19 and CCL21 levels in BALF. The interleukin (IL)-6 and tumor necrosis factor (TNF)-α levels in serum were also detected by ELISA. The protein expression levels of CCR7, as well as that of phosphorylated IκBα (p-IκBα) and phosphorylated p65 (p-p65) were determined by western blot analysis and RT-qPCR was used to determine the CCR7 mRNA levels. Our data demonstrated that the oral administration of baicalin significantly improved pulmonary function and attenuated inflammatory cell infiltration into the lungs. Baicalin also decreased the levels of OVA-IgE, IL-6, TNF-α and CCR7, as well as those of its ligand, CCL19; the levels of NF-κB were also markedly suppressed by baicalin. The CCR7 mRNA level was substantially decreased. Our results thus suggest that baicalin exerts an inhibitory effect on airway inflammation, and this effect may be associated with the inhibition of CCR7 and CCL19/CCL21, which may provide new mechanistic insight into the anti­inflammatory effects of baicalin.


Subject(s)
Asthma/prevention & control , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Flavonoids/pharmacology , Inflammation/prevention & control , NF-kappa B/metabolism , Receptors, CCR7/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/chemically induced , Asthma/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Female , Flavonoids/chemistry , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice, Inbred BALB C , Molecular Structure , Ovalbumin , Receptors, CCR7/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
12.
Environ Toxicol Pharmacol ; 46: 55-61, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27438894

ABSTRACT

Ligustrazine which is isolated from Chinese herb ligusticum chuanxiong hort, has been widely used in traditional Chinese medicine (TCM) for asthma treatment. In this study, we aim to observe the effect of ligustrazine on inflammation and the associated chemokines and receptors in ovalbumin (OVA)-induced mouse asthma model. Our data demonstrates that ligustrazine suppresses airway hyperresponsiveness to methacholine and lung inflammation in OVA-induced mouse asthma model. Ligustrazine also induces inhibition of inflammatory cells including neutrophils, lymphocytes and eosinophils. In addition, ligustrazine significantly reduces IL-4, IL-5, IL-17A, CCL3, CCL19 and CCL21 level in BALF of asthma mice. Furthermore, ligustrazine induces down-regulation of CCL19 receptor CCR7, STAT3 and p38 MAPK protein expression. Collectively, these results suggest that ligustrazine is effective in attenuation of allergic airway inflammatory changes and related chemokines and receptors in OVA-induced asthma model, and this action might be associated with inhibition of STAT3 and p38 MAPK pathway, which indicates that ligustrazine may be used as a potential therapeutic method to treat asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Chemokines/metabolism , Pyrazines/pharmacology , Animals , Asthma/chemically induced , Asthma/metabolism , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Female , Interleukins/metabolism , Lung/drug effects , Lung/pathology , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Ovalbumin/immunology , Ovalbumin/toxicity , Receptors, CCR7/metabolism , STAT3 Transcription Factor/metabolism
13.
Article in Chinese | WPRIM | ID: wpr-256595

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of low-selenium diet on the liver and kidneys of rats and explore the role of macrophage polarization into M1 and M2 phenotypes in liver and kidney injuries.</p><p><b>METHODS</b>Twenty-four rats (12 female and 12 male) were randomly divided into control group and low-selenium group and fed with normal chow (dietary selenium of 0.18 mg/kg) and low-selenium diet (dietary selenium of 0.02 mg/kg) for 109 days. After the feeding, the rats were sacrificed for HE staining to observe liver and kidney pathologies, and immunohistochemistry was performed for analyzing CCR7, CD206, CD163-positive cell numbers in the liver and kidneys.</p><p><b>RESULTS</b>The rats in low-selenium group showed severer fibrosis in the liver and kidney than the control group. In either male or female rats in low-selenium group, CCR7 and CD206 expressions in the liver were comparable with those in control group, but CD163 expression was lower than that in the control group (P<0.05 for both female and male rats). In the kidney, the proximal tubule showed a slightly higher while the distal tubule showed a slightly lower CCR7 expression in low selenium group than in the control group (P>0.05). In low-selenium group, a significantly lower CD163 expression in the distal tubule and a significantly higher CD206 expression in the proximal tubule were noted as compared with the control group (P<0.05 in both female and male rats). Compared with the control rats, the male rats in low-selenium group, but not the female rats, showed a significantly lower CD163 expression in the proximal tubule of the kidney (P<0.05); the female but not the male rats in low-selenium group show a higher CD206 expression in the distal tubule (P<0.05).</p><p><b>CONCLUSION</b>Low-selenium diet can cause liver and kidney fibrosis in rats and may inhibit macrophage activation into the M2 phenotype.</p>


Subject(s)
Animals , Female , Male , Rats , Antigens, CD , Metabolism , Antigens, Differentiation, Myelomonocytic , Metabolism , Diet , Fibrosis , Kidney , Metabolism , Pathology , Lectins, C-Type , Metabolism , Liver , Metabolism , Pathology , Macrophage Activation , Mannose-Binding Lectins , Metabolism , Receptors, CCR7 , Metabolism , Receptors, Cell Surface , Metabolism , Selenium
14.
Cell Mol Biol (Noisy-le-grand) ; 61(8): 37-43, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26667771

ABSTRACT

Berberine is an isoquinoline alkaloid found in several plant species like famous chinese herb, Rhizoma coptidis which has been used locally as a strong gastrointestinal remedy for thousands of years. The inhibitory effects of berberine on tumor progression properties have been reported before. In this study, we investigated the effect of berberine on an esophageal cancer cell line, KYSE-30 with emphasis on its effects on the expression of certain chemokine receptors. The cytotoxic effect of berberine on KYSE-30 cells was analyzed by MTT assay. In vitro cell migration assay was also applied to the treated cells and the expression levels of the selected chemokine receptors (CXCR4 and CCR7) was measured at mRNA level. A retarded growth, associated with increasing concentrations of berberine, was obvious. On the other hand, the migration rate of the cells was decreased when they were treated with different concentrations of berberine and the expression levels of the two chemokine receptors, involved in the migration and metastasis of esophageal cancer cells, were decreased following the same treatments. With these results, we tend to conclude that berberine might be a proper candidate for further investigations, by targeting the chemokine receptors, and possible applications as anti-metastatic agent in cancer studies.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Berberine/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Neoplastic , RNA, Messenger/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/isolation & purification , Berberine/isolation & purification , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/isolation & purification , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagus/drug effects , Esophagus/metabolism , Esophagus/pathology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR7/antagonists & inhibitors , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction
15.
Cell Physiol Biochem ; 35(2): 489-98, 2015.
Article in English | MEDLINE | ID: mdl-25613752

ABSTRACT

BACKGROUND/AIM: Recent studies have demonstrated that circulating fibrocytes contribute to the formation and development of fibrosis. Curcumin, a polyphenolic compound isolated from turmeric, has been shown to have anti-fibrotic effects in various organs. We and others have demonstrated that curcumin beneficially affects the development of fibrosis. However the effect of curcumin on circulating fibrocytes has not been reported. METHODS: Human circulating fibrocytes were isolated from leukocyte concentrates of healthy human donors and identified based on the expression of CD34, CD45, collagen I (COLI), and chemokine receptor CCR7 (CCR7) via flow cytometry. Cell Counting Kit-8 was used to evaluate cell viability. The effect of curcumin on the differentiation and migration of human circulating fibrocytes was evaluated by immunofluorescence staining, flow cytometry and a transwell migration assay. Transforming growth factor (TGF)-ß1 secretion was examined by ELISA. RESULTS: Curcumin treatment (72 h; 20 µM) significantly decreased the expression of COL I, α-SMA and CCR7, as well as TGF-ßl secretion, in human circulating fibrocytes. The inhibitory effect of curcumin on the differentiation and migration of human circulating fibrocytes is likely via regulating the CCR7/CCL21 signaling pathway, in particular by reducing CCR7 expression. These observed effects may be beneficial in resolving fibrosis by suppressing TGF-ß1 secretion. CONCLUSION: Our results suggest that curcumin has the potential to suppress the differentiation and migration of circulating fibrocytes, which would provide new explanation for curcumin's application in the development of fibrosis in various organs.


Subject(s)
Curcumin/pharmacology , Leukocytes/cytology , Leukocytes/drug effects , Receptors, CCR7/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Collagen Type I/metabolism , Down-Regulation , Fibrosis/drug therapy , Fibrosis/metabolism , Flow Cytometry , Humans , Leukocytes/metabolism , Transforming Growth Factor beta1/metabolism
16.
J Autoimmun ; 47: 58-72, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24075646

ABSTRACT

Autoimmunity is associated with a strong genetic component, but onset and persistence of clinically apparent autoimmune diseases often require an additional environmental trigger. The balance between immunity and tolerance is regulated by numerous molecular factors including nuclear hormone and homeostatic chemokine receptors. The nuclear hormone receptor RORγt and the chemokine receptor CCR7 are both essentially involved in functional lymphoid organogenesis and maintenance of lymphocyte homeostasis. Lack of one or the other impairs thymic T cell development and alters T cell homeostasis. Mice deficient for both, Ccr7(-/-)Rorγt(-/-), succumbed early to acute destructive inflammation, characterized by massive recruitment of inflammatory leukocytes, pro-inflammatory cytokine and autoantibody production, and wasting disease. Antibiotic-treatment of mice before disease onset reduced the overall gut microflora and abrogated the development of fatal mucosal inflammation. Hence, commensal bacteria and a confined tissue-specific inflammatory milieu serve as complementary trigger to initiate the lethal pathophysiologic process in Ccr7(-/-)Rorγt(-/-) mice.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity/immunology , Intestinal Mucosa/microbiology , Microbiota , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, CCR7/genetics , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Autoantibodies/immunology , Autoimmune Diseases/microbiology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Chimera/immunology , Colistin/therapeutic use , Inflammation/immunology , Intestinal Mucosa/immunology , Leukocytes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Streptomycin/therapeutic use
17.
Int J Hematol ; 96(6): 764-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23179902

ABSTRACT

The chemotactic movement of T lymphocytes mediated by chemokines and their receptors plays an important role in the pathogenesis of graft-versus-host disease (GVHD) post-allogeneic hematopoietic stem cell transplantation (allo-HSCT). CCR7 and CXCR3 are two receptors associated with the development of GVHD. Bortezomib, a proteasome inhibitor, was recently found to prevent GVHD in a mouse model and to decrease the production of Th1 cytokines. Here, we report that bortezomib differentially regulates the expression of CXCR3 and CCR7 on T cells; it significantly decreases CXCR3 expression on T cells as well as its CD4(+)/CD8(+) subsets in a dose-dependent manner, while it does not significantly affect CCR7 expression on T cells and subsets. Moreover, the secretion of CXCL9 by activated T cells is also increasingly downregulated with increasing concentrations of bortezomib. Meanwhile, bortezomib inhibits T-cell chemotactic movements toward CXCL9 in a dose-dependent manner, but has no effect on CCL19-induced T-cell chemotaxis. Additionally, it was found that bortezomib treatment also prompts T-lymphocyte apoptosis through activation of caspase-3 and its downstream PARP cleavage in a dose- and time-dependent manner. These results suggest that bortezomib may act as a suppressor of GVHD by downregulating T-cell chemotatic movement toward GVHD target organs, as well as by inducing apoptosis.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Chemokine CXCL9/metabolism , Chemotaxis/drug effects , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Receptors, CXCR3/biosynthesis , T-Lymphocyte Subsets/drug effects , Adult , Bortezomib , Cells, Cultured , Chemokine CCL19/physiology , Depression, Chemical , Down-Regulation , Drug Evaluation, Preclinical , Graft vs Host Disease/drug therapy , Humans , Lymphocyte Activation/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Protein Processing, Post-Translational , Receptors, CCR7/biosynthesis , Receptors, CCR7/genetics , Receptors, CXCR3/genetics , T-Lymphocyte Subsets/metabolism
18.
Rev Neurol ; 55(1): 31-7, 2012 Jul 01.
Article in Spanish | MEDLINE | ID: mdl-22718407

ABSTRACT

INTRODUCTION: Fingolimod has recently been approved for the therapy of relapsing multiple sclerosis. This drug binds to different sphingosine-1-phosphate receptors. AIM: To analyze basic mechanisms of action that can account for the efficacy of this drug in multiple sclerosis. DEVELOPMENT: Fingolimod acts as an inverse agonist on sphingosine-1-phosphate receptors, inducing degradation of receptors. On lymphoid circulation, this effect causes retention in lymph nodes of naive and central memory T cells, including Th17 T lymphocytes, bearing CCR7 and CD62L receptors. As a result, the level of circulating T cells is markedly decreased. B ell circulation is impaired and complex effects on other immune cells are also induced. Fingolimod enters the central nervous system and binds to receptors on glial cells and neurons. In experimental autoimmune encephalomyelitis, the therapeutic efficacy of fingolimod is not only associated with a reduced entry of inflammatory cells into the nervous system, but also with a direct effect mostly on astroglial cells. CONCLUSIONS: In multiple sclerosis patients, the available evidence indicates that fingolimod efficacy is directly associated with impairment of circulation of several T cell subsets and possibly B cells. Animal studies raise the possibility that an additional effect on glial cells might also contribute to the clinical efficacy.


Subject(s)
B-Lymphocytes/drug effects , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/drug therapy , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Th17 Cells/drug effects , Animals , Atrophy , B-Lymphocytes/immunology , Brain/immunology , Brain/pathology , Cell Movement , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/therapeutic use , L-Selectin/analysis , Lysophospholipids/physiology , Mice , Molecular Structure , Multiple Sclerosis/immunology , Neuroglia/drug effects , Neuroglia/immunology , Propylene Glycols/chemistry , Propylene Glycols/therapeutic use , Rats , Receptors, CCR7/analysis , Sphingosine/chemistry , Sphingosine/pharmacology , Sphingosine/physiology , Sphingosine/therapeutic use , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
19.
Lipids Health Dis ; 10: 203, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22070978

ABSTRACT

BACKGROUND: Long and persistent uncontrolled diabetes tends to degenerate the immune system and leads to an increased incidence of infection. Whey proteins (WPs) enhance immunity during early life and have a protective role in some immune disorders. In this study, the effects of camel WP on the chemotaxis of B and T cells to CXCL12 and CCL21 in diabetic mice were investigated. RESULTS: Flow cytometric analysis of the surface expressions of CXCR4 (CXCL12 receptor) and CCR7 (CCL21 receptor) on B and T cells revealed that the surface expressions of CXCR4 and CCR7 were not significantly altered in diabetic and WP-supplemented diabetic mice compared with control mice. Nevertheless, B and T lymphocytes from diabetic mice were found to be in a stunned state, with a marked and significant (P < 0.05) decrease in CXCL12- and CCL21-mediated actin polymerization and subsequently, a marked decrease in their chemotaxis. WP supplementation in the diabetes model was found to significantly increase CXCL12- and CCL21-mediated actin polymerization and chemotaxis in both B and T cells. CONCLUSION: Our data revealed the benefits of WP supplementation in enhancing cytoskeletal rearrangement and chemotaxis in B and T cells, and subsequently improving the immune response in diabetic mice.


Subject(s)
B-Lymphocytes/drug effects , Chemokine CCL21/metabolism , Chemokine CXCL12/metabolism , Chemotaxis/drug effects , Dietary Supplements , Milk Proteins/pharmacology , T-Lymphocytes/drug effects , Actins/metabolism , Animals , B-Lymphocytes/metabolism , Camelus , Cells, Cultured , Diabetes Mellitus, Experimental , Gene Expression , Male , Mice , Protein Multimerization/drug effects , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , T-Lymphocytes/metabolism , Whey Proteins
20.
Gastroenterology ; 141(6): 2130-2139.e11, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21875496

ABSTRACT

BACKGROUND & AIMS: The egress of memory T cells from peripheral tissues, such as lung and skin, into the draining lymph nodes requires their expression of CC chemokine receptor 7 (CCR7). In the intestine, resident memory T cells in the intestinal lamina propria (LP) do not express CCR7, indicating that they are tissue bound and do not exit the intestine. METHODS: We developed a cell transfer system, using rectal administration of lymphocytes to C57BL/6 mice. Lymphotoxin α-deficient mice were crossed with RAG-2(-/-) (recombination-activating gene-2) mice to generate lymphotoxin α-deficient × RAG-2(-/-) mice. RESULTS: Severe combined immunodeficient (SCID) or RAG-2(-/-) mice given rectal administration of splenic CD4(+) T cells from normal mice developed colitis; the cells proliferated not only in the LP but also in spleen. SCID or RAG-2(-/-) mice given rectal administrations of CD4(+) T cells that expressed green fluorescent protein (GFP(+)CD4(+) T cells) localized to the LP within 6 hours but were not found in the spleen until 24 hours after administration. Immunohistochemical and electron microscopic analyses detected CD4(+) T cells in the intraepithelial space just 3 hours after intrarectal administration. However, neither CCR7 deficiency nor the sphingosine-1-phosphate receptor agonist Fingolimod impaired the egress of CD4(+) T cells from LP to systemic circulation. CONCLUSIONS: CD4(+) T cells not only penetrate from the luminal side of the intestine to the LP but also actively egress from the LP into the circulation. We developed a rectal administration system that might be used to further investigate cell trafficking in intestinal mucosa and to develop enema-based therapeutics for intestinal diseases.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Cell Movement/physiology , Colitis, Ulcerative/immunology , Intestinal Mucosa/immunology , Mucous Membrane/immunology , Animals , Blood Circulation , Colitis, Ulcerative/blood , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, SCID , Microscopy, Electron , Receptors, CCR7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL