Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Inflammopharmacology ; 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753221

ABSTRACT

Inflammatory bowel diseases (IBDs) are prevalent and debilitating diseases with limited clinical treatment strategies. Mesenchymal stem cell (MSCs) are pluripotent stem cells with self-renewal capability and multiple immunomodulatory effects, which make them a promising therapeutic approach for IBDs. Thus, optimization of MSCs regimes is crucial for their further clinical application. Wogonin, a flavonoid-like compound with extensive immunomodulatory and adjuvant effects, has been investigated as a potential pretreatment for MSCs in IBD treatment. In this study, we employed the DSS-induced acute colitis mouse model to compare the therapeutic effectiveness of MSCs in pretreated with or without wogonin and further explore the underlying mechanism. Compared to untreated MSCs, MSCwogonin (pretreated with wogonin) showed greater effectiveness in the treatment of colitis. Further experiments revealed that wogonin treatment activated the AKT signaling pathway, resulting in higher cellular glycolysis. Inhibition of AKT phosphorylation by perifosine not only decreased glycolysis but impaired the therapeutic efficiency of MSCwogonin. Consistent with these results, qPCR data indicated that wogonin treatment induced the expression of immunomodulatory molecules IL-10, IDO, and AGR1, which were reduced by perifosine. Together, our data demonstrated that wogonin preconditioning strategy further augmented the therapeutic efficacy of MSCs via promoting glycolysis, which should be a promising strategy for optimizing MSCs therapy in IBDs.

2.
J Adv Res ; 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38070595

ABSTRACT

INTRODUCTION: Numerous studies demonstrated that NLRP3 has been implicated in the pathogenesis of inflammatory bowel disease (IBD). Mesenchymal stem cells (MSCs) regulated the NLRP3 inflammasome, which has emerged as a novel therapeutic approach for treating IBD. OBJECTIVES: The exact role of NLRP3 in regulating MSCs' function is unclear. Our study aimed to explore how NLRP3 affects the therapeutic effects of MSCs in colitis. METHODS: We extracted MSCs from the bone marrow of C57BL/6 mice and Nlrp3 KO mice, and identified them using differentiation assays and flow cytometry. In vitro, Both WT MSCs and Nlrp3 KO MSCs were stimulated with inflammatory factor Lipopolysaccharide (LPS), and only WT MSCs were stimulated with varying concentrations of the NLRP3 inhibitor MCC950, then, quantified IL-10 levels in the supernatant. RNA-seq was performed to examine gene expression patterns and Seahorse was used to assess oxidative phosphorylation (OXPHOS) and glycolysis levels. Western blot was used to evaluate protein expression. In vivo, we treated DSS-induced colitis with either WT or Nlrp3 KO MSCs, monitoring weight, measuring colon length, and further evaluation. We also treated DSS-induced colitis with pretreated MSCs (BAY876, oe-Glut1, or oe-NLRP3), following the same experimental procedures as described above. RESULTS: Our results demonstrate that Nlrp3 deletion did not affect MSC phenotypes, but rather promoted osteogenic differentiation. However, the absence of Nlrp3 reduced IL-10 production in MSCs in the presence of LPS, leading to impaired protection on DSS-induced colitis. Conversely, overexpression of NLRP3 promotes the production of IL-10, enhancing therapeutic effects. Further investigation revealed that Nlrp3 deficiency downregulated Glut1 expression and glycolysis activation in MSCs, resulting in decreased IL-10 production. Notably, overexpressing Glut1 in Nlrp3 KO MSCs restored their therapeutic effect that was previously dampened due to Nlrp3 deletion. CONCLUSION: Our findings demonstrate that NLRP3 heightens the therapeutic effects of MSC treatment on DSS-induced colitis.

3.
Adv Sci (Weinh) ; 10(27): e2302640, 2023 09.
Article in English | MEDLINE | ID: mdl-37485650

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide. Patient-derived organoid (PDO) has great potential in precision oncology, but low success rate, time-consuming culture, and lack of tumor microenvironment (TME) limit its application. Mesenchymal stromal cells (MSC) accumulate in primary site to support tumor growth and recruit immune cells to form TME. Here, MSC and peripheral blood mononuclear cells (PBMC) coculture is used to construct HCC organoid-on-a-chip mimicking original TME and provide a high-throughput drug-screening platform to predict outcomes of anti-HCC immunotherapies. HCC-PDOs and PBMC are co-cultured with MSC and Cancer-associated fibroblasts (CAF). MSC increases success rate of biopsy-derived PDO culture, accelerates PDO growth, and promotes monocyte survival and differentiation into tumor-associated macrophages. A multi-layer microfluidic chip is designed to achieve high-throughput co-culture for drug screening. Compared to conventional PDOs, MSC-PDO-PBMC and CAF-PDO-PBMC models show comparable responses to chemotherapeutic or targeted anti-tumor drugs but more precise prediction potential in assessing patients' responses to anti-PD-L1 drugs. Moreover, this microfluidic platform shortens PDO growth time and improves dimensional uniformity of organoids. In conclusion, the study successfully constructs microengineered organoid-on-a-chip to mimic TME for high-throughput drug screening, providing novel platform to predict immunotherapy response of HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mesenchymal Stem Cells , Humans , Carcinoma, Hepatocellular/therapy , Leukocytes, Mononuclear , Liver Neoplasms/therapy , Precision Medicine , Organoids , Immunotherapy , Lab-On-A-Chip Devices , Tumor Microenvironment
6.
Immunology ; 167(4): 576-589, 2022 12.
Article in English | MEDLINE | ID: mdl-35899990

ABSTRACT

Regulatory B cells (Bregs) are immune cells that constrain autoimmune response and restrict inflammation via their expression of interleukin (IL)-10. However, the molecular mechanisms underlying Breg differentiation and IL-10 secretion remain unclear. Previous data suggest that cellular metabolism determines both the fate and function of these cells. Here, we suggest an essential role for mitochondrial oxidative phosphorylation (OXPHOS) in the regulation of IL-10 in these Bregs. We found that IL-10+ B cells from IL-10-green fluorescent protein-expressing mice had higher oxygen consumption rate than IL-10- B cells. In addition, inhibition of OXPHOS decreased the expression of IL-10 in B cells. Furthermore, suppression of OXPHOS diminished the expression of surface markers for Bregs and impaired their therapeutic effects in dextran sulphate sodium (DSS)-induced colitis. Mechanistically, mitochondrial OXPHOS was found to regulate the transcription factor HIF-1α through the extracellular signal-related kinase pathway. Taken together, this study reveals a strong correlation between mitochondrial OXPHOS and Breg phenotype/function, indicating OXPHOS as a therapeutic target in autoimmune diseases driven by Breg dysfunction.


Subject(s)
B-Lymphocytes, Regulatory , Colitis , Mice , Animals , Interleukin-10/genetics , Interleukin-10/metabolism , Oxidative Phosphorylation , Mice, Inbred C57BL
7.
Front Cell Dev Biol ; 9: 762669, 2021.
Article in English | MEDLINE | ID: mdl-34722547

ABSTRACT

Proper development of mammalian skeletal muscle relies on precise gene expression regulation. Our previous studies revealed that muscle development is regulated by both mRNA and long non-coding RNAs (lncRNAs). Accumulating evidence has demonstrated that N6-methyladenosine (m6A) plays important roles in various biological processes, making it essential to profile m6A modification on a transcriptome-wide scale in developing muscle. Patterns of m6A methylation in lncRNAs in developing muscle have not been uncovered. Here, we reveal differentially expressed lncRNAs and report temporal m6A methylation patterns in lncRNAs expressed in mouse myoblasts and myotubes by RNA-seq and methylated RNA immunoprecipitation (MeRIP) sequencing. Many lncRNAs exhibit temporal differential expression, and m6A-lncRNAs harbor the consensus m6A motif "DRACH" along lncRNA transcripts. Interestingly, we found that m6A methylation levels of lncRNAs are positively correlated with the transcript abundance of lncRNAs. Overexpression or knockdown of m6A methyltransferase METTL3 alters the expression levels of these lncRNAs. Furthermore, we highlight that the function of m6A genic lncRNAs might correlate to their nearby mRNAs. Our work reveals a fundamental expression reference of m6A-mediated epitranscriptomic modifications in lncRNAs that are temporally expressed in developing muscle.

8.
Front Cell Dev Biol ; 9: 744171, 2021.
Article in English | MEDLINE | ID: mdl-34660602

ABSTRACT

N6-methyladenosine (m6A) RNA methylation has emerged as an important factor in various biological processes by regulating gene expression. However, the dynamic profile, function and underlying molecular mechanism of m6A modification during skeletal myogenesis remain elusive. Here, we report that members of the m6A core methyltransferase complex, METTL3 and METTL14, are downregulated during skeletal muscle development. Overexpression of either METTL3 or METTL14 dramatically blocks myotubes formation. Correspondingly, knockdown of METTL3 or METTL14 accelerates the differentiation of skeletal muscle cells. Genome-wide transcriptome analysis suggests ERK/MAPK is the downstream signaling pathway that is regulated to the greatest extent by METTL3/METTL14. Indeed, METTL3/METTL14 expression facilitates ERK/MAPK signaling. Via MeRIP-seq, we found that MNK2, a critical regulator of ERK/MAPK signaling, is m6A modified and is a direct target of METTL3/METTL14. We further revealed that YTHDF1 is a potential reader of m6A on MNK2, regulating MNK2 protein levels without affecting mRNA levels. Furthermore, we discovered that METTL3/14-MNK2 axis was up-regulated notably after acute skeletal muscle injury. Collectively, our studies revealed that the m6A writers METTL3/METTL14 and the m6A reader YTHDF1 orchestrate MNK2 expression posttranscriptionally and thus control ERK signaling, which is required for the maintenance of muscle myogenesis and may contribute to regeneration.

9.
Mol Ther Nucleic Acids ; 26: 222-236, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34513306

ABSTRACT

Mesenchymal stromal cells (MSCs) have shown significant heterogeneity in terms of therapeutic efficacy for inflammatory bowel disease (IBD) treatment, which may be due to an insufficient number of MSCs homing to the damaged tissue of the colon. Engineering MSCs with specific chemokine receptors can enhance the homing ability by lentiviral transduction. However, the unclear specific chemokine profile related to IBD and the safety concerns of viral-based gene delivery limit its application. Thus, a new strategy to modify MSCs to express specific chemokine receptors using mRNA engineering is developed to evaluate the homing ability of MSCs and its therapeutic effects for IBD. We found that CXCL2 and CXCL5 were highly expressed in the inflammatory colon, while MSCs minimally expressed the corresponding receptor CXCR2. Transient expression of CXCR2 in MSC was constructed and exhibited significantly enhanced migration to the inflamed colons, leading to a robust anti-inflammatory effect and high efficacy. Furthermore, the high expression of semaphorins7A on MSCs were found to induce the macrophages to produce IL-10, which may play a critical therapeutic role. This study demonstrated that the specific chemokine receptor CXCR2 mRNA-engineered MSCs not only improves the therapeutic efficacy of IBD but also provides an efficient and safe MSC modification strategy.

10.
Ocul Surf ; 22: 103-109, 2021 10.
Article in English | MEDLINE | ID: mdl-34333155

ABSTRACT

PURPOSE: To investigate the safety and feasibility of topical injection of bone marrow derived mesenchymal stem cells (BM-MSCs) in acute severe ocular burns. METHODS: In this open-label,single-arm study, subconjunctival injection of allogenic BM-MSCs combined with standard treatment was given to 16 patients with acute severe ocular burns (Dua's grade IV to VI) within 2 weeks after injury. The primary outcome was efficacy rate which referred to the proportion of complete corneal epithelialization patients without perforation. The secondary outcome was safety, visual acuity, the number of symblephara, and elevated intraocular pressure. RESULTS: One patient was lost to follow-up. During the follow-up period, no corneal perforation was developed. Complete corneal epithelialization was noted 8 (ranged 4-10 weeks) weeks after treatment in 13 eyes (81.3%). The efficacy rate was 87.5% (95% confidence interval, CI: 61.7-98.4). Hypopyon was detected and later well controlled in 1 eye. Partial or total limbal stem cell deficiency (LSCD) was noted in all eyes. Improvement of visual acuity was achieved in 5 out of 16 eyes (31.3%). Seven eyes' visual acuity was reached 0.1. Symblepharon with varied severity was noted in 5 eyes. Two eyes had elevated intraocular pressure. CONCLUSIONS: This study confirms the safety of subconjunctival injection of BM-MSCs as an innovative and convenient procedure in ocular burns. The overall result is promising considering the absence of perforation, the low severity of symblepharon and visual acuity improvement.


Subject(s)
Burns, Chemical , Corneal Diseases , Eye Burns , Limbus Corneae , Mesenchymal Stem Cells , Burns, Chemical/therapy , Corneal Diseases/therapy , Eye Burns/chemically induced , Eye Burns/therapy , Feasibility Studies , Humans , Retrospective Studies
11.
Front Immunol ; 12: 662441, 2021.
Article in English | MEDLINE | ID: mdl-34248942

ABSTRACT

Objective: To investigate the efficacy and safety of bone marrow-derived mesenchymal stem cells (BM-MSCs) on chronic active antibody-mediated rejection (cABMR) in the kidney allograft. Methods: Kidney recipients with biopsy-proven cABMR were treated with allogeneic third-party BM-MSCs in this open-label, single-arm, single-center, two-dosing-regimen phase I/II clinical trial. In Regimen 1 (n=8), BM-MSCs were administered intravenously at a dose of 1.0×106 cells/kg monthly for four consecutive months, while in Regimen 2 (n=15), the BM-MSCs dose was 1.0×106 cells/kg weekly during four consecutive weeks. The primary endpoints were the absolute change of estimated glomerular filtration rate (eGFR) from baseline (delta eGFR) and the incidence of adverse events associated with BM-MSCs administration 24 months after the treatment. Contemporaneous cABMR patients who did not receive BM-MSCs were retrospectively analyzed as the control group (n =30). Results: Twenty-three recipients with cABMR received BM-MSCs. The median delta eGFR of the total BM-MSCs treated patients was -4.3 ml/min per 1.73m2 (interquartile range, IQR -11.2 to 1.2) 2 years after BM-MSCs treatment (P=0.0233). The median delta maximum donor-specific antibody (maxDSA) was -4310 (IQR -9187 to 1129) at 2 years (P=0.0040). The median delta eGFR of the control group was -12.7 ml/min per 1.73 m2 (IQR -22.2 to -3.5) 2 years after the diagnosis, which was greater than that of the BM-MSCs treated group (P=0.0342). The incidence of hepatic enzyme elevation, BK polyomaviruses (BKV) infection, cytomegalovirus (CMV) infection was 17.4%, 17.4%, 8.7%, respectively. There was no fever, anaphylaxis, phlebitis or venous thrombosis, cardiovascular complications, or malignancy after BM-MSCs administration. Flow cytometry analysis showed a significant decreasing trend of CD27-IgD- double negative B cells subsets and trend towards the increase of CD3+CD4+PD-1+/lymphocyte population after MSCs therapy. Multiplex analysis found TNF-α, CXCL10, CCL4, CCL11 and RANTES decreased after MSCs treatment. Conclusion: Kidney allograft recipients with cABMR are tolerable to BM-MSCs. Immunosuppressive drugs combined with intravenous BM-MSCs can delay the deterioration of allograft function, probably by decreasing DSA level and reducing DSA-induced injury. The underlying mechanism may involve immunomodulatory effect of MSCs on peripheral B and T cells subsets.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Graft Rejection/immunology , Graft Rejection/prevention & control , Kidney Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Transplantation, Homologous/adverse effects , Adolescent , Adult , Aged , Antibodies , Bone Marrow Cells/immunology , Female , Humans , Immunomodulation , Immunosuppressive Agents/therapeutic use , Kidney/pathology , Male , Middle Aged , Retrospective Studies , Young Adult
12.
Oxid Med Cell Longev ; 2021: 5527935, 2021.
Article in English | MEDLINE | ID: mdl-34239686

ABSTRACT

Inflammatory bowel diseases (IBD) are prevalent and debilitating diseases; their clinical remedy is desperately unmet. Mesenchymal stem cells (MSCs) are pluripotent stem cells with multiple immunomodulatory effects, which are attributed to their efficacy in the IBD rodent model. Optimization of MSC regimes in IBD is a crucial step for their further clinical application. Wogonin is a flavonoid-like compound, which showed extensive immunomodulatory and adjuvant effects. This research is aimed at investigating whether and how Wogonin boosted the therapeutic efficiency of MSCs on DSS-induced colitis. Our results showed that the MSC treatment with Wogonin significantly alleviated the intestinal inflammation in IBD mice by increased IL-10 expression. In vitro experiments, Wogonin obviously raised the IL-10 production and ROS levels of MSCs in a dose-dependent manner. Meanwhile, western blot data suggested Wogonin improves the IL-10 production by inducing transcript factor HIF-1α expression via AKT/GSK3ß signal pathway. Finally, the favorable effects of Wogonin on MSCs were confirmed by IL-10 blockade experiment in vivo. Together, our results suggested that Wogonin significantly increased the IL-10 production and enhanced the therapeutic effects of MSCs in DSS-induced colitis. This work suggested Wogonin as a novel optimal strategy for MSC clinical application.


Subject(s)
Colitis/chemically induced , Colitis/therapy , Drugs, Chinese Herbal/therapeutic use , Flavanones/therapeutic use , Interleukin-10/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Dextran Sulfate/adverse effects , Drugs, Chinese Herbal/pharmacology , Flavanones/pharmacology , Male , Mice
13.
Huan Jing Ke Xue ; 42(8): 3904-3912, 2021 Aug 08.
Article in Chinese | MEDLINE | ID: mdl-34309277

ABSTRACT

To evaluate the pollution by polycyclic aromatic hydrocarbons (PAHs) in the surface soil of the core urban area of Lanzhou, 62 topsoil samples were collected from the area. The soil samples were analyzed for the content of 16 priority PAHs, using gas chromatography-mass spectrometry. Descriptive statistical methods were used to characterize contamination by PAHs. An absolute principal component analysis-multiple linear regression (APCS-MLR) model was applied to determine the sources of PAHs in the soil samples, and the accuracy of the model results was verified. Finally, the main influence regions of each source were determined with a geo-statistical method. The results showed that the contents of Σ16 PAHs in the surface soils of Lanzhou ranged from 1069 to 7377 µg ·kg-1, with an average of 2423 µg ·kg-1. High molecular weight PAHs (4-6 rings) were dominant, accounting for 72.81% of the Σ16 PAHs. Verification results of the APCS-MLR model showed that the measured values were in good correspondence with the predicted values, which indicated that the model had good applicability for source apportionment of soil PAHs in the study area. The main sources of PAHs in Lanzhou were traffic emissions (35.42%), petroleum emissions (29.88%), and a mixture of coal and biomass combustion (33.91%). The sources were greatly affected by human activities, and high values were mainly distributed in traffic-intensive and industrial areas. The results indicated that the sources of soil PAHs were complex and influenced by anthropogenic activities in the study area. Stringent control measures should be placed on the sources and areas of influence that contribute to soil PAHs to reduce the emissions and the level of soil pollution resulting from PAHs.


Subject(s)
Polycyclic Aromatic Hydrocarbons , Soil Pollutants , China , Coal/analysis , Environmental Monitoring , Environmental Pollution , Geographic Information Systems , Humans , Linear Models , Polycyclic Aromatic Hydrocarbons/analysis , Risk Assessment , Soil , Soil Pollutants/analysis
14.
Stem Cells Dev ; 30(4): 203-213, 2021 02.
Article in English | MEDLINE | ID: mdl-33371825

ABSTRACT

Heart transplantation is the final life-saving therapeutic strategy for many end-stage heart diseases. Long-term immunosuppressive regimens are needed to prevent allograft rejection. Mesenchymal stromal cells (MSCs) have been shown as immunomodulatory therapy for organ transplantation. However, the effect of dose and timing of MSC treatment on heart transplantation has not yet been examined. In this study, we infused three doses (1 × 106, 2 × 106, or 5 × 106 cells) of human MSCs (hMSCs) to the recipient BALB/c mice before (7 days or 24 h) or after (24 h) receiving C57BL/6 cardiac transplants. We found that infusion of high dose hMSCs (5 × 106) at 24 h post-transplantation significantly prolonged the survival time of cardiac grafts. To delineate the underlying mechanism, grafts, spleens, and draining lymph nodes were harvested for analysis. Dose-dependent effect of hMSC treatment was shown in: (1) alleviation of International Society of Heart and Lung Transplantation (ISHLT) score in grafts; (2) reduction of the population of CD4+ and CD8+ T cells; (3) increase of regulatory T (Treg) cells; (4) and decrease of serum levels of inflammatory cytokines and donor-specific antibodies. Taken together, we showed timing critical and dose-dependent immunomodulatory effects of hMSC treatment against acute allograft rejection in a mouse model of heart transplantation.


Subject(s)
Graft Rejection/therapy , Heart Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Allografts , Animals , Cytokines/blood , Cytokines/immunology , Flow Cytometry , Graft Rejection/etiology , Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation/adverse effects , Humans , Kaplan-Meier Estimate , Male , Mesenchymal Stem Cells/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , Time Factors , Transplantation Tolerance/immunology , Transplantation, Heterologous
15.
J Neuroinflammation ; 17(1): 347, 2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33213462

ABSTRACT

BACKGROUND: The R1441G mutation in the leucine-rich repeat kinase 2 (LRRK2) gene results in late-onset Parkinson's disease (PD). Peripheral inflammation and gut microbiota are closely associated with the pathogenesis of PD. Chronic periodontitis is a common type of peripheral inflammation, which is associated with PD. Porphyromonas gingivalis (Pg), the most common bacterium causing chronic periodontitis, can cause alteration of gut microbiota. It is not known whether Pg-induced dysbiosis plays a role in the pathophysiology of PD. METHODS: In this study, live Pg were orally administrated to animals, three times a week for 1 month. Pg-derived lipopolysaccharide (LPS) was used to stimulate mononuclear cells in vitro. The effects of oral Pg administration on the gut and brain were evaluated through behaviors, morphology, and cytokine expression. RESULTS: Dopaminergic neurons in the substantia nigra were reduced, and activated microglial cells were increased in R1441G mice given oral Pg. In addition, an increase in mRNA expression of tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß) as well as protein level of α-synuclein together with a decrease in zonula occludens-1 (Zo-1) was detected in the colon in Pg-treated R1441G mice. Furthermore, serum interleukin-17A (IL-17A) and brain IL-17 receptor A (IL-17RA) were increased in Pg-treated R1441G mice. CONCLUSIONS: These findings suggest that oral Pg-induced inflammation may play an important role in the pathophysiology of LRRK2-associated PD.


Subject(s)
Gastrointestinal Microbiome/physiology , Immunity/physiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/immunology , Microglia/immunology , Neurodegenerative Diseases/immunology , Porphyromonas gingivalis/immunology , Administration, Oral , Animals , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/immunology , Cells, Cultured , Dopaminergic Neurons/immunology , Dopaminergic Neurons/microbiology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Mice, Transgenic , Microglia/microbiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/microbiology , Permeability , Substantia Nigra/immunology , Substantia Nigra/microbiology
16.
Immunopharmacol Immunotoxicol ; 42(5): 473-483, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32806961

ABSTRACT

OBJECTIVES: Amphiregulin (Areg), a glycoprotein from the epidermal growth factor receptor (EGFR) ligand family, has a well-documented protective role against tissue injury; however, its effects on immune-mediated liver injury are still unclear. Here, we used a concanavalin A (ConA)-induced acute liver hepatitis model to explore the effects of Areg on immune-mediated acute liver injury. MATERIALS AND METHODS: Some C57BL/6 mice were administered ConA at a dose of 20 mg/kg (model mice), and some received 5 µg of Areg (treated mice). Then, their survival rates over 36 h were analyzed. After 5 h of treatment, liver function, hepatic histology, and apoptosis in liver tissue were investigated, and cytokine expression and neutrophil infiltration and activity in the liver were detected. Moreover, the protective effects of Areg were also evaluated without IL-22 in vivo. RESULTS: Our results showed that Areg administration increased acute liver failure (ALF) mouse survival, restored liver function, and alleviated liver damage. Interestingly, Areg administration increased IL-22 production in hepatic T cells and upregulated IL-22 concentrations in the serum and liver, whereas IL-22 neutralization completely abolished the therapeutic effect of Areg. Meanwhile, Areg administration was concomitant with increased expression of the anti-apoptotic proteins Bcl-2 and Bcl-xL, which are important in the hepatoprotective mechanism of IL-22. CONCLUSIONS: Areg showed direct protective effects against ConA-induced acute liver injury, which suggests the potential therapeutic application of Areg in immune-mediated ALF.


Subject(s)
Amphiregulin/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Interleukins/metabolism , Liver Failure, Acute/prevention & control , Liver/drug effects , T-Lymphocytes/drug effects , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Concanavalin A , Disease Models, Animal , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/immunology , Liver Failure, Acute/metabolism , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , bcl-X Protein/metabolism , Interleukin-22
17.
J Inflamm Res ; 13: 377-386, 2020.
Article in English | MEDLINE | ID: mdl-32801831

ABSTRACT

BACKGROUND AND PURPOSE: Both NLRP3 inflammasome and chemokines are involved in the initiation and development of acute lung inflammation, but the underlying mechanism is still elusive. The present study investigated the role of chemokines and NLRP3 in recruiting neutrophils in the early phase of acute lung injury. METHODS: In an endotoxin (lipopolysaccharide [LPS])-induced acute lung injury model, we measured the lung injury severity, myeloperoxidase (MPO) activity and chemokine profiles in wild-type (WT) and NLRP3 knockout (NLRP3-/-) mice, and then identified the key chemokines by specific antibody blockage. RESULTS: The results showed that NLRP3 deficiency was associated with alleviating lung damage, by reducing alveolar epithelial cell apoptosis and decreasing neutrophil accumulation. Furthermore, compared with WT mice, IL-1ß, CCL2, CXCL1, CXCL5 and CXCL12 levels from the serum of NLRP3-/- mice were much lower after exposure to LPS. However, in lung tissue, only lower CXCL12 levels were observed from the NLRP3-/- ALI mice, and higher levels of CXCR4 were expressed in NLRP3-/- neutrophils. Blockage of CXCL12 dramatically relieved the severity of ALI and reduced neutrophil accumulation in the lung. CONCLUSION: NLRP3 alters CXCL12 expression in acute lung injury. CXCL12 is crucial for neutrophil recruitment in NLRP3-mediated neutrophilic lung injury.

18.
J Immunol Res ; 2020: 3032425, 2020.
Article in English | MEDLINE | ID: mdl-32566686

ABSTRACT

Wogonin (5,7-dihydroxy-8-methoxyflavone) is an ingredient of the extracts from Scutellaria baicalensis, which has documented a wide spectrum of anti-inflammatory and antitumor activities, including inhibiting regulatory T cells, regulating effector T cell functions, and mediating macrophage immunity. However, the potential effect of Wogonin on B cells has not been fully understood. Here, our results showed that Wogonin inhibited IL-10 secretion in B cells. When purified B cells were activated by lipopolysaccharide (LPS) in vitro, the amount of IL-10 production in supernatant was decreased by Wogonin significantly. The protective role of B cells on dextran sulfate sodium- (DSS-) induced colitis was alleviated after exposure to Wogonin. Furthermore, administration of Wogonin on LPS-treated B cells suppressed phosphorylation of STAT3 and ERK, but not AKT. Interestingly, among those IL-10 signaling-associated transcription factors, mRNA and protein levels of Hif-1α were specifically decreased by Wogonin. Overall, our study indicates that Wogonin suppresses potentially IL-10 production in B cells via inhibition of the STAT3 and ERK signaling pathway as well as inhibition of mRNA and protein levels of the transcription factor Hif-1α. These results provide novel and potential molecular targets of Wogonin in B cells and help us further understand its mechanism of action, which could potentially improve its clinical application in the future.


Subject(s)
B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Drugs, Chinese Herbal/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavanones/pharmacology , Interleukin-10/biosynthesis , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Cytokines/biosynthesis , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Immunophenotyping , Male , Mice , Transcription, Genetic
19.
Theranostics ; 9(16): 4633-4647, 2019.
Article in English | MEDLINE | ID: mdl-31367246

ABSTRACT

Rationale: Mesenchymal stem cells (MSCs) have been demonstrated to ameliorate inflammatory bowel disease by their actions on multiple immune cells, especially on regulatory B cells (Breg cells). However, the phenotypes and functions of human MSCs (hMSCs)-treated Breg cell subsets are not yet clear. Methods: Purified B cells were cocultured with MSCs and the phenotypes and immunomodulatory functions of the B cells were analyzed by FACS and proliferation assays in vitro. Also, a trinitrobenzenesulfonic acid-induced mouse colitis model was employed to detect the function of MSC-treated Breg cells in vivo. Results: We demonstrated that coculturing with hMSCs significantly enhanced the immunomodulatory activity of B cells by up-regulating IL-10 expression. We then identified that a novel regulatory B cell population characterized by CD23 and CD43 phenotypic markers could be induced by hMSCs. The CD23+CD43+ Breg cells substantially inhibited the inflammatory cytokine secretion and proliferation of T cells through an IL-10-dependent pathway. More significantly, intraperitoneal injection of hMSCs ameliorated the clinical and histopathological severity in the mouse experimental colitis model, accompanied by an increase in the number of CD23+CD43+ Breg cells. The adoptive transfer of CD23+CD43+ B cells effectively alleviated murine colitis, as compared with the CD23-CD43- B cells. Treatment with CD23+CD43+ B cells, and not hMSCs, substantially improved the symptoms of colitis in B cell-depleted mice. Conclusion: the novel CD23+CD43+ Breg cell subset appears to be involved in the immunomodulatory function of hMSCs and sheds new light on elucidating the therapeutic mechanism of hMSCs for the treatment of inflammation-related diseases.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Colitis/therapy , Immunomodulation , Inflammation/therapy , Inflammatory Bowel Diseases/therapy , Mesenchymal Stem Cells/immunology , Animals , Cell Proliferation , Coculture Techniques , Colitis/chemically induced , Colitis/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Interleukins/immunology , Leukosialin/immunology , Mice , Mice, Inbred BALB C , Receptors, IgE/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...