Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 576
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731942

ABSTRACT

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


Subject(s)
Acinar Cells , Carcinoma, Pancreatic Ductal , Metaplasia , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Reactive Oxygen Species , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , Reactive Oxygen Species/metabolism , Humans , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Metaplasia/metabolism , Metaplasia/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Mice, Transgenic , Chemokines, CC/metabolism , Chemokines, CC/genetics , Macrophage Inflammatory Proteins/metabolism , Macrophage Inflammatory Proteins/genetics , Pancreas/metabolism , Pancreas/pathology
2.
Am J Respir Cell Mol Biol ; 68(2): 176-185, 2023 02.
Article in English | MEDLINE | ID: mdl-36174229

ABSTRACT

Tissue availability remains an important limitation of single-cell genomic technologies for investigating cellular heterogeneity in human health and disease. BAL represents a minimally invasive approach to assessing an individual's lung cellular environment for diagnosis and research. However, the lack of high-quality, healthy lung reference data is a major obstacle to using single-cell approaches to study a plethora of lung diseases. Here, we performed single-cell RNA sequencing on over 40,000 cells isolated from the BAL of four healthy volunteers. Of the six cell types or lineages we identified, macrophages were consistently the most numerous across individuals. Our analysis confirmed the expression of marker genes defining cell types despite background signals because of the ambient RNA found in many single-cell studies. We assessed the variability of gene expression across macrophages and defined a distinct subpopulation of cells expressing a set of genes associated with Macrophage Inflammatory Protein 1 (MIP-1). RNA in situ hybridization and reanalysis of published lung single-cell data validated the presence of this macrophage subpopulation. Thus, our study characterizes lung macrophage heterogeneity in healthy individuals and provides a valuable resource for future studies to understand the lung environment in health and disease.


Subject(s)
Macrophage Inflammatory Proteins , Macrophages , Humans , Macrophage Inflammatory Proteins/genetics , Bronchoalveolar Lavage Fluid , Healthy Volunteers , RNA
3.
Shock ; 58(2): 179-188, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35953456

ABSTRACT

ABSTRACT: Sex-related outcome differences in trauma remain controversial. The mechanisms causing sex-biased outcomes are likely to have hormonal and genetic components, in which X-linked genetic polymorphisms may play distinct roles because of X-linked inheritance, hemizygosity in males, and X chromosome mosaicism in females. The study aimed to elucidate the contribution of biological sex and the common X-linked IRAK1 haplotype to posttrauma clinical complications, inflammatory cytokine and chemokine production, and polymorphonuclear cell and monocyte activation. Postinjury clinical outcome was tested in 1507 trauma patients (1,110 males, 397 females) after stratification by sex or the variant IRAK1 haplotype. Males showed a three- to fivefold greater frequency of posttrauma sepsis, but similar mortality compared to females. Stratification by the variant IRAK1 haplotype revealed increased pneumonia and urinary tract infection in Wild type (WT) versus variant IRAK1 males, whereas increased respiratory failures in variant versus WT females. Cytokine/chemokine profiles were tested in whole blood from a subset of patients (n = 81) and healthy controls (n = 51), which indicated sex-related differences in ex vivo lipopolysaccharide responsiveness manifesting in a 1.5- to 2-fold increased production rate of tumor necrosis factor α, interleukin-1ß (IL-1ß), IL-10, Macrophage Inflammatory Protein-1 Alpha, and MIP1ß in WT male compared to WT female trauma patients. Variant IRAK1 decreased IL-6, IL-8, and interferon gamma-induced protein 10 production in male trauma subjects compared to WT, whereas cytokine/chemokine responses were similar in variant IRAK1 and WT female trauma subjects. Trauma-induced and lipopolysaccharide-stimulated polymorphonuclear cell and monocyte activation determined by using a set of cluster of differentiation markers and flow cytometry were not influenced by sex or variant IRAK1. These findings suggest that variant IRAK1 is a potential contributor to sex-based outcome differences, but its immunomodulatory impacts are modulated by biological sex.


Subject(s)
Genes, X-Linked , Interleukin-10 , Antigens, CD , Cytokines/genetics , Female , Haplotypes/genetics , Humans , Interferon-gamma , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1beta , Interleukin-6 , Interleukin-8 , Lipopolysaccharides , Macrophage Inflammatory Proteins/genetics , Male , Phenotype , Tumor Necrosis Factor-alpha/genetics
4.
AIDS ; 36(9): 1215-1222, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35608113

ABSTRACT

OBJECTIVE: Despite lower plasma HIV RNA levels, women progress faster to AIDS than men. The reasons for these differences are not clear but might be a consequence of an elevated inflammatory response in women. METHODS: We investigated sex differences in cytokine profiles by measuring the concentrations of 36 cytokines/chemokines by Luminex in blood of women and men (sex at birth) with chronic HIV infection under suppressive therapy. We initially performed a principal component analysis to see if participants clustered by sex, and then fit a partial least squares discriminant analysis (PLS-DA) model where we used cytokines to predict sex at birth. The significance of the difference in nine cytokines with VIP greater than 1 was tested using Wilcoxon test-rank. Further, potential confounding factors were tested by multivariate linear regression models. RESULTS: Overall, we predicted sex at birth in the PLS-DA model with an error rate of approximately 13%. We identified five cytokines, which were significantly higher in women compared with men, namely the pro-inflammatory chemokines CXCL1 (Gro-α), CCL5 (RANTES), CCL3 (MIP-1α), CCL4 (MIP-1ß), as well as the T-cell homeostatic factor IL-7. The effect of sex remained significant after adjusting for CD4 + , age, ethnicity, and race for all cytokines, except for CCL3 and race. CONCLUSION: The observed sex-based differences in cytokines might contribute to higher immune activation in women compared with men despite suppressive therapy. Increased levels of IL-7 in women suggest that homeostatic proliferation may have a differential contribution to HIV reservoir maintenance in female and male individuals. Our study emphasizes the importance of sex-specific studies of viral pathogenesis.


Subject(s)
Anti-Retroviral Agents , Cytokines , HIV Infections , Sex Characteristics , Anti-Retroviral Agents/therapeutic use , Chemokine CCL4 , Chemokine CCL5/analysis , Chemokine CCL5/genetics , Chemokines , Cytokines/blood , Female , HIV Infections/drug therapy , Humans , Interleukin-7 , Macrophage Inflammatory Proteins/genetics , Male
5.
Signal Transduct Target Ther ; 6(1): 91, 2021 02 28.
Article in English | MEDLINE | ID: mdl-33640900

ABSTRACT

Eosinophils are terminally differentiated cells derived from hematopoietic stem cells (HSCs) in the bone marrow. Several studies have confirmed the effective roles of eosinophils in asthmatic airway pathogenesis. However, their regulatory functions have not been well elucidated. Here, increased C-C chemokine ligand 6 (CCL6) in asthmatic mice and the human orthologs CCL15 and CCL23 that are highly expressed in asthma patients are described, which are mainly derived from eosinophils. Using Ccl6 knockout mice, further studies revealed CCL6-dependent allergic airway inflammation and committed eosinophilia in the bone marrow following ovalbumin (OVA) challenge and identified a CCL6-CCR1 regulatory axis in hematopoietic stem cells (HSCs). Eosinophil differentiation and airway inflammation were remarkably decreased by the specific CCR1 antagonist BX471. Thus, the study identifies that the CCL6-CCR1 axis is involved in the crosstalk between eosinophils and HSCs during the development of allergic airway inflammation, which also reveals a potential therapeutic strategy for targeting G protein-coupled receptors (GPCRs) for future clinical treatment of asthma.


Subject(s)
Asthma/genetics , Chemokines, CC/genetics , Eosinophils/metabolism , Macrophage Inflammatory Proteins/genetics , Receptors, CCR1/genetics , Adolescent , Adult , Aged , Animals , Asthma/pathology , Bone Marrow/drug effects , Cell Differentiation/drug effects , Eosinophils/pathology , Female , Healthy Volunteers , Hematopoietic Stem Cells/metabolism , Humans , Hypersensitivity/genetics , Hypersensitivity/pathology , Inflammation/genetics , Inflammation/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Middle Aged , Ovalbumin/pharmacology , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Signal Transduction/drug effects , Young Adult
6.
Sci Rep ; 11(1): 3786, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33589722

ABSTRACT

Protein-bound uremic toxins (Indoxyl sulfate [IS] and p-cresyl sulfate [PCS]) are both associated with cardiovascular (CV) and all-cause mortality in subjects with chronic kidney disease (CKD). Possible mechanisms have not been elucidated. In hemodialysis patients, we investigated the relationship between the free form of IS and PCS and 181 CV-related proteins. First, IS or PCS concentrations were checked, and high levels were associated with an increased risk of acute coronary syndrome (ACS) in 333 stable HD patients. CV proteins were further quantified by a proximity extension assay. We examined associations between the free form protein-bound uremic toxins and the quantified proteins with correction for multiple testing in the discovery process. In the second step, the independent association was evaluated by multivariable-adjusted models. We rank the CV proteins related to protein-bound uremic toxins by bootstrapped confidence intervals and ascending p-value. Six proteins (signaling lymphocytic activation molecule family member 5, complement component C1q receptor, C-C motif chemokine 15 [CCL15], bleomycin hydrolase, perlecan, and cluster of differentiation 166 antigen) were negatively associated with IS. Fibroblast growth factor 23 [FGF23] was the only CV protein positively associated with IS. Three proteins (complement component C1q receptor, CCL15, and interleukin-1 receptor-like 2) were negatively associated with PCS. Similar findings were obtained after adjusting for classical CV risk factors. However, only higher levels of FGF23 was related to increased risk of ACS. In conclusion, IS and PCS were associated with several CV-related proteins involved in endothelial barrier function, complement system, cell adhesion, phosphate homeostasis, and inflammation. Multiplex proteomics seems to be a promising way to discover novel pathophysiology of the uremic toxin.


Subject(s)
Cresols/adverse effects , Indican/adverse effects , Renal Insufficiency, Chronic/drug therapy , Sulfuric Acid Esters/adverse effects , Toxins, Biological/chemistry , Acute Coronary Syndrome/chemically induced , Acute Coronary Syndrome/genetics , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Chemokines, CC/genetics , Cresols/administration & dosage , Cysteine Endopeptidases/genetics , Female , Fibroblast Growth Factor-23/genetics , Heparan Sulfate Proteoglycans/genetics , Humans , Indican/administration & dosage , Macrophage Inflammatory Proteins/genetics , Male , Middle Aged , Protein Binding/drug effects , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/genetics , Signaling Lymphocytic Activation Molecule Family/genetics , Sulfuric Acid Esters/administration & dosage , Toxins, Biological/adverse effects , Toxins, Biological/genetics
7.
Mol Med Rep ; 22(6): 5339-5347, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33174007

ABSTRACT

Wound healing is a complex physiological process in which fibrocytes serve a vital role. However, the mechanism underlying the recruitment of fibrocytes during wound healing remains largely unknown. The present study aimed to investigate whether endothelial cells are involved in the recruitment of fibrocytes in wound healing. To mimic the in vivo angiogenic process, a co­culture system consisting of endothelial cells and fibrocytes was achieved using a permeable Transwell co­culture system. The expression of chemokines produced by endothelial cells with or without co­culture was then measured using a gene chip. Based on the dataset from chip analysis, chemokine ligand 15 (CCL15) produced by endothelial cells was identified, which likely serves a regulatory role in mediating the transmigration of fibrocytes. Overexpression of CCL15 in endothelial cells or chemokine receptor 1 (CCR1) in fibrocytes promoted the transmigration of fibrocytes, whilst silencing the expression of CCL15 in endothelial cells or that of CCR1 in fibrocytes attenuated the transmigration of fibrocytes. Results from the present study suggested that the CCL15­CCR1 axis between endothelial cells and fibrocytes serves a vital role in mediating the recruitment of fibrocytes during wound healing.


Subject(s)
Chemokines, CC/metabolism , Macrophage Inflammatory Proteins/metabolism , Monocytes/metabolism , Receptors, CCR1/metabolism , Cell Line, Tumor , Cell Movement , Chemokines/metabolism , Chemokines, CC/genetics , Chemokines, CC/physiology , Coculture Techniques/methods , Endothelial Cells/metabolism , Humans , Ligands , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/physiology , Mesenchymal Stem Cells/metabolism , Receptors, CCR1/physiology , Receptors, Chemokine/metabolism , Wound Healing/physiology
8.
Biomed Pharmacother ; 129: 110380, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32554250

ABSTRACT

Baoyuan Jiedu (BYJD for short) decoction, a traditional Chinese medicine formula, is composed of Astragalus, Ginseng, Aconite root, Honeysuckle, Angelica, Licorice, which has the functions of nourishing qi and blood, enhancing immune function, improving quality of life and prolonging survival time of tumor patients. The present study aimed to investigate the effect and mechanism of BYJD decoction on reversing the pre-metastatic niche. We showed that BYJD decoction could prolong the survival time of 4T1 tumor-bearing mice. Moreover, we found that the BYJD decoction inhibited the formation of lung pre-metastatic niche and inhibited recruitment of myeloid derived suppressor cells (MDSCs) in the lung. Mechanistically, we showed that the proteins and genes expression of TGF-ß, Smad2, Smad3, p-Smad2/3, Smad4, CCL9 in the TGF-ß/CCL9 signaling pathway were suppressed by BYJD decoction. In line with the above findings, our results confirm that BYJD decoction inhibits the accumulation of MDSC in pre-metastatic niche of lung via TGF-ß/CCL9 pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Chemokines, CC/metabolism , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/prevention & control , Lung/drug effects , Macrophage Inflammatory Proteins/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chemokines, CC/genetics , Female , Gene Expression Regulation, Neoplastic , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophage Inflammatory Proteins/genetics , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Signal Transduction , Transforming Growth Factor beta/genetics , Tumor Burden/drug effects
9.
Immunobiology ; 225(3): 151911, 2020 05.
Article in English | MEDLINE | ID: mdl-32059938

ABSTRACT

A deep knowledge of the profound immunological response induced by traumatic brain injury (TBI) raises the possibility of novel therapeutic interventions. Existing studies have highlighted the important roles of C-C motif ligands in the development of neuroinflammation after brain injury; however, the participation of macrophage inflammatory protein-1 (MIP-1) family members in this phenomenon is still undefined. Therefore, the goal of our study was to evaluate changes in macrophage inflammatory protein-1 (MIP-1) family members (CCL3, CCL4, and CCL9) and their receptors (CCR1 and CCR5) in a mouse model of TBI (induced by controlled cortical impact (CCI)). We also investigated the pattern of activation of immunological cells (such as neutrophils, microglia and astroglia), which on one hand express CCR1/CCR5, and on the other hand might be a source of the tested chemokines in the injured brain. We investigated changes in mRNA (RT-qPCR) and/or protein (ELISA and Western blot) expression in brain structures (the cortex, hippocampus, thalamus, and striatum) at different time points (24 h, 4 days, 7 days, 2 weeks, and/or 5 weeks) after trauma. Our time-course studies revealed the upregulation of the mRNA expression of all members of the MIP-1 family (CCL3, CCL4, and CCL9) in all tested brain structures, mainly in the early stages after injury. A similar pattern of activation was observed at the protein level in the cortex and thalamus, where the strongest activation was observed 1 day after CCI; however, we did not observe any change in CCL3 in the thalamus. Analyses of CCR1 and CCR5 demonstrated the upregulation of the mRNA expression of both receptors in all tested cerebral structures, mainly in the early phases post injury (24 h, 4 days and 7 days). Protein analysis showed the upregulation of CCR1 and CCR5 in the thalamus 24 h after TBI, but we did not detect any change in the cortex. We also observed the upregulation of neutrophil marker (MPO) at the early time points (24 h and 7 days) in the cortex, while the profound activation of microglia (IBA-1) and astroglia (GFAP) was observed mainly on day 7. Our findings highlight for the first time that CCL3, CCL4, CCL9 and their receptors offer promising targets for influencing secondary neuronal injury and improving TBI therapy. The results suggest that the MIP-1 family is an important target for pharmacological intervention for brain injury.


Subject(s)
Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/metabolism , Gene Expression Regulation , Macrophage Inflammatory Proteins/genetics , Multigene Family , Animals , Biomarkers , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Macrophage Inflammatory Proteins/metabolism , Mice , Microglia/metabolism , Neurons/metabolism , Neutrophils/immunology , Neutrophils/metabolism
10.
Theranostics ; 9(18): 5122-5133, 2019.
Article in English | MEDLINE | ID: mdl-31410205

ABSTRACT

Controlling bilirubin to a low level is necessary in physiology because of its severe neurotoxicity. Therefore, it is of great interest to understand the regulatory mechanisms for bilirubin homeostasis. In this study, we uncover a critical role for circadian clock in regulation of bilirubin detoxification and homeostasis. Methods: The mRNA and protein levels of Bmal1 (a core clock gene), metabolic enzymes and transporters were measured by qPCR and Western blotting, respectively. Luciferase reporter, mobility shift and chromatin immunoprecipitation were used to investigate transcriptional gene regulation. Experimental hyperbilirubinemia was induced by injection of bilirubin or phenylhydrazine. Unconjugated bilirubin (UCB) and conjugated bilirubin were assessed by ELISA. Results: We first demonstrated diurnal variations in plasma UCB levels and in main bilirubin-detoxifying genes Ugt1a1 and Mrp2. Of note, the circadian UCB levels were antiphase to the circadian expressions of Ugt1a1 and Mrp2. Bmal1 ablation abrogated the circadian rhythms of UCB and bilirubin-induced hepatotoxicity in mice. Bmal1 ablation also decreased mRNA and protein expressions of both Ugt1a1 and Mrp2 in mouse livers, and blunted their circadian rhythms. A combination of luciferase reporter, mobility shift, and chromatin immunoprecipitation assays revealed that Bmal1 trans-activated Ugt1a1 and Mrp2 through specific binding to the E-boxes in the promoter region. Further, Bmal1 ablation caused a loss of circadian time-dependency in bilirubin clearance and sensitized mice to chemical induced-hyperbilirubinemia. Moreover, bilirubin stimulated Bmal1 expression through antagonism of Rev-erbα, constituting a feedback mechanism in bilirubin detoxification. Conclusion: These data supported a dual role for circadian clock in regulation of bilirubin detoxification, generating circadian variations in bilirubin level via direct transactivation of detoxifying genes Ugt1a1 and Mrp2, and defending the body against hyperbilirubinemia via Rev-erbα antagonism. Thereby, our study provided a potential mechanism for management of bilirubin related diseases.


Subject(s)
ARNTL Transcription Factors/genetics , Bilirubin/metabolism , Circadian Clocks/genetics , Feedback, Physiological , Hyperbilirubinemia/genetics , ARNTL Transcription Factors/metabolism , Animals , Cell Line , Chemokines, CC/genetics , Chemokines, CC/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Inactivation, Metabolic/genetics , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Mice, Inbred C57BL , Transcription, Genetic
11.
J Innate Immun ; 11(1): 63-73, 2019.
Article in English | MEDLINE | ID: mdl-30189430

ABSTRACT

BACKGROUND: We have previously identified the upregulation of the innate immune response, neutrophil activation, and apoptosis during anaphylaxis using a microarray approach. This study aimed to validate the differential gene expression and investigate protein concentrations of "hub genes" and upstream regulators during anaphylaxis. METHODS: Samples were collected from patients with anaphylaxis on their arrival at the emergency department, and after 1 and 3 h. mRNA levels of 11 genes (interleukin-6 [IL-6], IL-10, oncostatin M [OSM], S100A8, S100A9, matrix metalloproteinase 9 [MMP9], FASL, toll-like receptor 4 [TLR4], MYD88, triggering receptor expressed on myeloid cells 1 [TREM1], and cluster of differentiation 64 [CD64]) were measured in peripheral blood leucocytes using qPCR. Serum protein concentrations were measured by ELISA or cytometric bead array for 6 of these candidates. RESULTS: Of 69 anaphylaxis patients enrolled, 36 (52%) had severe reactions, and 38 (55%) were female. Increases in both mRNA and protein of IL-10, S100A9, MMP9, and TREM1 were observed. OSM, S100A8, TLR4, and CD64 were upregulated and IL-6 protein concentrations were increased during anaphylaxis. Both FASL and soluble Fas ligand decreased during anaphylaxis. CONCLUSION: These results provide evidence for the involvement of innate immune pathways and myeloid cells during human anaphylaxis, validating previous microarray findings. Elevated S100A8, S100A9, TLR4, and TREM1 expression, and increased S100A9 and soluble TREM1 protein concentrations strongly suggest that neutrophils are activated during acute anaphylaxis.


Subject(s)
Anaphylaxis/immunology , Immunity, Innate , Myeloid Cells/immunology , Neutrophil Activation , Neutrophils/immunology , Adult , Anaphylaxis/blood , Anaphylaxis/genetics , Cytokines/genetics , Cytokines/metabolism , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Gene Expression Profiling , Humans , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , Young Adult
12.
Int J Mol Sci ; 19(7)2018 Jul 23.
Article in English | MEDLINE | ID: mdl-30041454

ABSTRACT

Although nano-copper is currently used extensively, the adverse effects on liver cytochrome P450 (CYP450) enzymes after oral exposure are not clear. In this study, we determined the effects and mechanisms of action of nano- and micro-copper on the expression and activity of CYP450 enzymes in rat liver. Rats were orally exposed to micro-copper (400 mg/kg), Cu ion (100 mg/kg), or nano-copper (100, 200 and 400 mg/kg) daily for seven consecutive days. Histopathological, inflammatory and oxidative stress were measured in the livers of all rats. The mRNA levels and activity of CYP450 enzymes, as well as the mRNA levels of select nuclear receptors, were determined. Exposure to nano-copper (400 mg/kg) induced significant oxidative stress and inflammation relative to the controls, indicated by increased levels of interleukin (IL)-2, IL-6, interferon (IFN)-γ, macrophage inflammatory protein (MIP-1), total antioxidant capacity (T-AOC), malondialdehyde (MDA), inducible nitric oxide synthase (iNOS) and nitric oxide (NO) after exposure. The levels of mRNA expression of pregnane X receptor (PXR), constitutive androstane receptor (CAR) and aryl hydrocarbon receptor (AHR) were significantly decreased in 400 mg/kg nano-copper treated rats. Nano-copper activated the expression of the NF-kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT)3 signaling pathways. Nano-copper decreased the mRNA expression and activity of CYP 1A2, 2C11, 2D6, 2E1 and 3A4 in a dose-dependent manner. The adverse effects of micro-copper are less severe than those of nano-copper on the CYP450 enzymes of rats after oral exposure. Ingestion of large amounts of nano-copper in animals severely affects the drug metabolism of the liver by inhibiting the expression of various CYP450 enzymes, which increases the risk of drug-drug interactions in animals.


Subject(s)
Copper/adverse effects , Cytochrome P-450 Enzyme System/metabolism , Liver/drug effects , Metal Nanoparticles/adverse effects , Animals , Copper/chemistry , Cytochrome P-450 Enzyme System/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Liver/metabolism , MAP Kinase Signaling System , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Male , Malondialdehyde/metabolism , Metal Nanoparticles/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pregnane X Receptor , Rats , Rats, Sprague-Dawley , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
13.
Infect Genet Evol ; 64: 126-130, 2018 10.
Article in English | MEDLINE | ID: mdl-29902581

ABSTRACT

It is known that multiple genetic variants can affect immune responses to the hepatitis B virus (HBV) vaccine. A case-control study was undertaken to examine the possible association of low responsiveness to the HBV vaccine in a Chinese population with genetic polymorphisms in integrin subunit alpha L, CD58, tumor necrosis factor superfamily member 15, C-C motif chemokine ligand 15, transforming growth factor beta 3, and B-cell lymphoma 6 protein. The copy numbers of these six genes were detected in 129 low responders, 129 middle responders and 129 high responders to HBV vaccination. There were no significant differences in the copy numbers of these six genes between the groups. Thus, these findings indicated that the copy number variations of these genes may not be the reason for the low responsiveness to the HBV vaccine in a Chinese population.


Subject(s)
Asian People/genetics , Genetic Variation , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B/genetics , Hepatitis B/immunology , CD58 Antigens/genetics , Chemokines, CC/genetics , Disease Susceptibility , Female , Gene Dosage , Hepatitis B/prevention & control , Humans , Macrophage Inflammatory Proteins/genetics , Male , Proto-Oncogene Proteins c-bcl-6/genetics , Selection, Genetic , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
14.
Blood ; 131(15): 1743-1754, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29348127

ABSTRACT

Improved diagnostic and treatment methods are needed for chronic graft-versus-host disease (cGVHD), the leading cause of late nonrelapse mortality (NRM) in long-term survivors of allogenic hematopoietic cell transplantation. Validated biomarkers that facilitate disease diagnosis and classification generally are lacking in cGVHD. Here, we conducted whole serum proteomics analysis of a well-established murine multiorgan system cGVHD model. We discovered 4 upregulated proteins during cGVHD that are targetable by genetic ablation or blocking antibodies, including the RAS and JUN kinase activator, CRKL, and CXCL7, CCL8, and CCL9 chemokines. Donor T cells lacking CRK/CRKL prevented the generation of cGVHD, germinal center reactions, and macrophage infiltration seen with wild-type T cells. Whereas antibody blockade of CCL8 or CXCL7 was ineffective in treating cGVHD, CCL9 blockade reversed cGVHD clinical manifestations, histopathological changes, and immunopathological hallmarks. Mechanistically, elevated CCL9 expression was present predominantly in vascular smooth muscle cells and uniquely seen in cGVHD mice. Plasma concentrations of CCL15, the human homolog of mouse CCL9, were elevated in a previously published cohort of 211 cGVHD patients compared with controls and associated with NRM. In a cohort of 792 patients, CCL15 measured at day +100 could not predict cGVHD occurring within the next 3 months with clinically relevant sensitivity/specificity. Our findings demonstrate for the first time the utility of preclinical proteomics screening to identify potential new targets for cGVHD and specifically CCL15 as a diagnosis marker for cGVHD. These data warrant prospective biomarker validation studies.


Subject(s)
Chemokines, CC/blood , Graft vs Host Disease/blood , Macrophage Inflammatory Proteins/blood , Proteome/metabolism , Animals , Biomarkers/blood , Chemokines, CC/genetics , Chronic Disease , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Macrophage Inflammatory Proteins/genetics , Mice , Proteome/genetics , Proteomics
15.
Clin Cancer Res ; 23(3): 833-844, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27492974

ABSTRACT

PURPOSE: We have reported loss of SMAD4 promotes expression of CCL15 from colorectal cancer to recruit CCR1+ myeloid cells through the CCL15-CCR1 axis, which contributes to invasion and liver metastasis. However, the molecular mechanism of lung metastasis is yet to be elucidated. Our purpose is to determine whether similar mechanism is involved in the lung metastasis of colorectal cancer. EXPERIMENTAL DESIGN: In a mouse model, we examined whether SMAD4 could affect the metastatic activity of colorectal cancer cells to the lung through the CCL15-CCR1 axis. We immunohistochemically analyzed expression of SMAD4, CCL15, and CCR1 with 107 clinical specimens of colorectal cancer lung metastases. We also characterized the CCR1+ myeloid cells using several cell-type-specific markers. RESULTS: In a mouse model, CCL15 secreted from SMAD4-deficient colorectal cancer cells recruited CCR1+ cells, promoting their metastatic activities to the lung. Immunohistochemical analysis of lung metastases from colorectal cancer patients revealed that CCL15 expression was significantly correlated with loss of SMAD4, and that CCL15-positive metastases recruited approximately 1.9 times more numbers of CCR1+ cells than CCL15-negative metastases. Importantly, patients with CCL15-positive metastases showed a significantly shorter relapse-free survival (RFS) than those with CCL15-negative metastases, and multivariate analysis indicated that CCL15 expression was an independent predictor of shorter RFS. Immunofluorescent staining showed that most CCR1+ cells around lung metastases were tumor-associated neutrophil, although a minor fraction was granulocytic myeloid-derived suppressor cell. CONCLUSIONS: CCL15-CCR1 axis may be a therapeutic target to prevent colorectal cancer lung metastasis. CCL15 can be a biomarker indicating poor prognosis of colorectal cancer patients with lung metastases. Clin Cancer Res; 23(3); 833-44. ©2016 AACR.


Subject(s)
Chemokines, CC/physiology , Colorectal Neoplasms/pathology , Lung Neoplasms/secondary , Macrophage Inflammatory Proteins/physiology , Neoplasm Proteins/deficiency , Neutrophil Infiltration , Receptors, CCR1/physiology , Smad4 Protein/deficiency , Animals , Cell Line, Tumor , Cell Movement , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Colorectal Neoplasms/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Heterografts , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Nude , Mice, SCID , Myeloid Cells/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Prognosis , Proportional Hazards Models , Smad4 Protein/physiology
16.
BMC Cancer ; 16: 98, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26875556

ABSTRACT

BACKGROUND: The differential diagnosis of follicular thyroid carcinoma (FTC) and follicular adenoma (FA) before surgery is a clinical challenge. Many efforts have been made but most focusing on tumor cells, while the roles of tumor associated macrophages (TAMs) remained unclear in FTC. Here we analyzed the differences between TAMs in FTC and those in FA. METHODS: We first analyzed the density of TAMs by CD68 immunostaining in 59 histologically confirmed FTCs and 47 FAs. Cytokines produced by FTC and FA were profiled using antibody array, and validated by quantitative PCR. Chemotaxis of monocyte THP-1 was induced by condition medium of FTC cell lines (FTC133 and WRO82-1) with and without anti-CCL15 neutralizing antibody. Finally, we analyzed CCL15 protein level in FTC and FA by immunohistochemistry. RESULTS: The average density of CD68(+) cells was 9.5 ± 5.4/field in FTC, significantly higher than that in FA (4.9 ± 3.4/field, p < 0.001). Subsequently profiling showed that CCL15 was the most abundant chemokine in FTC compared with FA. CCL15 mRNA in FTC was 51.4-folds of that in FA. CM of FTC cell lines induced THP-1 cell chemotaxis by 33 ~ 77%, and anti-CCL15 neutralizing antibody reduced THP-1 cell migration in a dose-dependent manner. Moreover, we observed positive CCL15 immunostaining in 67.8% of FTCs compared with 23.4% of FAs. CONCLUSION: Our study suggested FTC might induce TAMs infiltration by producing CCL15. Measurement of TAMs and CCL15 in follicular thyroid lesions may be applied clinically to differentiate FTC from FA pre-operation.


Subject(s)
Adenocarcinoma, Follicular/diagnosis , Adenoma/diagnosis , Chemokines, CC/biosynthesis , Diagnosis, Differential , Macrophage Inflammatory Proteins/biosynthesis , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Adenoma/genetics , Adenoma/pathology , Biopsy, Fine-Needle , Chemokines, CC/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophage Inflammatory Proteins/genetics , Macrophages/pathology , Male , Preoperative Period , RNA, Messenger/biosynthesis , Tissue Array Analysis
17.
Stem Cells ; 34(4): 1112-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26763650

ABSTRACT

Mesenchymal stem cells (MSCs) possess the ability to migrate toward tumor sites and are regarded as promising gene delivery vehicles for cancer therapeutics. However, the factors that mediate this tropism have yet to be completely elucidated. In this study, through cytokine array analysis, chemokine CCL15 was found to be the most abundant protein differentially expressed in hepatocellular carcinoma (HCC) cell lines compared with a normal liver cell line. Serum CCL15 levels in HCC patients determined by enzyme linked immunosorbent assay were shown to be profoundly elevated compared with healthy controls. Immunohistochemical analysis indicated that CCL15 expression was much stronger in HCC tumor tissues than in adjacent nontumor tissues. Transwell migration assay suggested that CCL15 may be involved in chemotaxis of human MSCs (hMSCs) toward HCC in vitro and that this chemotactic effect of CCL15 is mediated via CCR1 receptors on hMSCs. Orthotopic animal models of HCC were established to investigate the role of CCL15 in hMSCs migration toward HCC in vivo. Both histological and flow cytometric analysis showed that significantly fewer hMSCs localized within 97H-CCL15-shRNA xenografts compared with 97H-green fluorescent protein xenografts after intravenous delivery. Finally, the possible effects of hMSCs on HCC tumor growth were also evaluated. Coculture experiments showed that hMSCs had no apparent effect on the proliferation of HCC cells in vitro In addition, systemic administration of hMSCs did not affect HCC tumor progression in vivo. Our data in this study help to elucidate the mechanism underlying the homing capacity of hMSCs toward HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Chemokines, CC/genetics , Gene Transfer Techniques , Liver Neoplasms/therapy , Macrophage Inflammatory Proteins/genetics , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Chemokines, CC/biosynthesis , Chemokines, CC/therapeutic use , Chemotaxis/genetics , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Humans , Liver Neoplasms/genetics , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/therapeutic use , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/cytology , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Receptors, CCR1/biosynthesis , Receptors, CCR1/genetics , Xenograft Model Antitumor Assays
18.
Oncotarget ; 6(35): 37012-27, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26473289

ABSTRACT

Sertoli cells, the primary somatic cell in the seminiferous epithelium, provide the spermatogonial stem cell (SSC) microenvironment (niche) through physical support and the expression of paracrine factors. However, the regulatory mechanisms within the SSC niche, which is primarily controlled by Sertoli cells, remain largely unknown. GATA4 is a Sertoli cell marker, involved in genital ridge initiation, sex determination and differentiation during the embryonic stage. Here, we showed that neonatal mice with a targeted disruption of Gata4 in Sertoli cells (Gata4(flox/flox); Amh-Cre; hereafter termed Gata4 cKO) displayed a loss of the establishment and maintenance of the SSC pool and apoptosis of both gonocyte-derived differentiating spermatogonia and meiotic spermatocytes. Thus, progressive germ cell depletion and a Sertoli-cell-only syndrome were observed as early as the first wave of murine spermatogenesis. Transplantation of germ cells from postnatal day 5 (P5) Gata4 cKO mice into Kit(W/W-v) recipient seminiferous tubules restored spermatogenesis. In addition, microarray analyses of P5 Gata4 cKO mouse testes showed alterations in chemokine signaling factors, including Cxcl12, Ccl3, Cxcr4 (CXCL12 receptor), Ccr1 (CCL3 receptor), Ccl9, Xcl1 and Ccrl2. Deletion of Gata4 in Sertoli cells markedly attenuated Sertoli cell chemotaxis, which guides SSCs or prospermatogonia to the stem cell niche. Finally, we showed that GATA4 transcriptionally regulated Cxcl12 and Ccl9, and the addition of CXCL12 and CCL9 to an in vitro testis tissue culture system increased the number of PLZF+ undifferentiated spermatogonia within Gata4 cKO testes. Together, these results reveal a novel role for GATA4 in controlling the SSC niche via the transcriptional regulation of chemokine signaling shortly after birth.


Subject(s)
Chemokine CXCL12/metabolism , Chemokines, CC/metabolism , GATA4 Transcription Factor/physiology , Macrophage Inflammatory Proteins/metabolism , Sertoli Cells/pathology , Spermatogonia/pathology , Stem Cell Niche , Stem Cells/pathology , Animals , Animals, Newborn , Blotting, Western , Cell Differentiation , Cells, Cultured , Chemokine CXCL12/genetics , Chemokines, CC/genetics , Chemotaxis , Female , Gene Expression Regulation, Developmental , Humans , Luciferases/metabolism , Macrophage Inflammatory Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/metabolism , Signal Transduction , Spermatogenesis , Spermatogonia/metabolism , Stem Cells/metabolism
19.
Sci Rep ; 5: 10453, 2015 May 28.
Article in English | MEDLINE | ID: mdl-26020416

ABSTRACT

This study aimed to explore whether and how L-cystathionine had any regulatory effect on the inflammatory response in THP-1-derived macrophages cultured in vitro under oxidized low-density lipoprotein (ox-LDL) stimulation. The human monocyte line THP-1 cell was cultured in vitro and differentiated into macrophages after 24 hours of PMA induction. Macrophages were pretreated with L-cystathionine and then treated with ox-LDL. The results showed that compared with the controls, ox-LDL stimulation significantly upregulated the expression of THP-1-derived macrophage MCP-1 by enhancing NF-κB p65 phosphorylation, nuclear translocation and DNA binding with the MCP-1 promoter. Compared with the ox-LDL group, 0.3 mmol/L and 1.0 mmol/L L-cystathionine significantly inhibited the expression of THP-1-derived macrophage MCP-1. Mechanistically, 0.3 mmol/L and 1.0 mmol/L L-cystathionine suppressed phosphorylation and nuclear translocation of the NF-κB p65 protein, as well as the DNA binding activity and DNA binding level of NF-κB with the MCP-1 promoter, which resulted in a reduced THP-1-derived macrophage MCP-1 generation. This study suggests that L-cystathionine could inhibit the expression of MCP-1 in THP-1-derived macrophages induced by ox-LDL via inhibition of NF-κB p65 phosphorylation, nuclear translocation, and binding of the MCP-1 promoter sequence after entry into the nucleus.


Subject(s)
Macrophage Inflammatory Proteins/genetics , Macrophages/metabolism , Transcription Factor RelA/biosynthesis , Cell Differentiation/drug effects , Cystathionine/administration & dosage , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/metabolism , Macrophage Inflammatory Proteins/metabolism , Macrophages/drug effects , Macrophages/pathology , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation/drug effects , Transcription Factor RelA/genetics
20.
J Virol ; 89(10): 5502-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25740989

ABSTRACT

UNLABELLED: Elite controllers (ECs) are a rare group of HIV seropositive individuals who are able to control viral replication without antiretroviral therapy. The mechanisms responsible for this phenotype, however, have not been fully elucidated. In this study, we examined CD4(+) T cell resistance to HIV in a cohort of elite controllers and explored transcriptional signatures associated with cellular resistance. We demonstrate that a subgroup of elite controllers possess CD4(+) T cells that are specifically resistant to R5-tropic HIV while remaining fully susceptible to X4-tropic and vesicular stomatitis virus G (VSV-G)-pseudotyped viruses. Transcriptome analysis revealed 17 genes that were differentially regulated in resistant elite controllers relative to healthy controls. Notably, the genes encoding macrophage inflammatory protein 1α (MIP-1α), CCL3 and CCL3L1, were found to be upregulated. The MIP-1α, MIP-1ß, and RANTES chemokines are natural ligands of CCR5 and are known to interfere with HIV replication. For three elite controllers, we observed increased production of MIP-1α and/or MIP-1ß at the protein level. The supernatant from resistant EC cells contained MIP-1α and MIP-1ß and was sufficient to confer R5-tropic resistance to susceptible CD4(+) T cells. Additionally, this effect was reversed by using inhibitory anti-MIP antibodies. These results suggest that the T cells of these particular elite controllers may be naturally resistant to HIV infection by blocking R5-tropic viral entry. IMPORTANCE: HIV is a pandemic health problem, and the majority of seropositive individuals will eventually progress to AIDS unless antiretroviral therapy (ART) is administered. However, rare patients, termed elite controllers, have a natural ability to control HIV infection in the absence of ART, but the mechanisms by which they achieve this phenotype have not been fully explored. This paper identifies one mechanism that may contribute to this natural resistance: some elite controllers have CD4(+) T cells that produce high levels of MIP chemokines, which block R5-tropic HIV entry. This mechanism could potentially be exploited to achieve a therapeutic effect in other HIV-seropositive individuals.


Subject(s)
HIV Infections/immunology , HIV Long-Term Survivors , HIV-1 , Macrophage Inflammatory Proteins/blood , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Chemokine CCL3/blood , Chemokine CCL3/genetics , Chemokine CCL4/blood , Chemokine CCL4/genetics , Chemokine CCL5/blood , Chemokine CCL5/genetics , Chemokines, CC/blood , Chemokines, CC/genetics , Cohort Studies , Female , Gene Dosage , HIV Infections/genetics , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Host-Pathogen Interactions , Humans , Macrophage Inflammatory Proteins/genetics , Male , Middle Aged , RNA, Messenger/blood , RNA, Messenger/genetics , Receptors, CCR5/blood , Receptors, CXCR4/blood , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...