Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Eur J Pharm Sci ; 199: 106805, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38763450

ABSTRACT

Drug resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is a primary factor affecting their therapeutic efficacy in human non-small cell lung cancer (NSCLC). NSCLC cells can undergo epithelial-mesenchymal transition (EMT) induced by many factors in the tumour microenvironment (TME), which plays a crucial role in tumour drug resistance. In this study, a multicellular lung-on-a-chip that can realise the cell co-culture of the human non-small cell lung cancer cell line HCC827, human foetal lung fibroblasts (HFL-1), and human umbilical vein endothelial cells (HUVECs) is prepared. The TME was simulated on the chip combined with perfusion and other factors, and the drug evaluation of osimertinib was performed to explore the drug resistance mechanism of EGFR-TKIs. In the early stages, a two-dimensional static cell co-culture was achieved by microchip, and the results showed that HFL-1 cells could be transformed into cancer-associated fibroblasts (CAFs), and HCC827 cells could undergo EMT, both of which were mediated by Interleukin-6 (IL-6). Vimentin (VIM) and Alpha Skeletal Muscle Actin (a-SMA) expression of HFL-1 was upregulated, whereas E-cadherin (E-cad) expression of HCC827 was down-regulated. Further, N-cadherin (N-cad) expression of HCC827 was upregulated. In both the static cell co-culture and multicellular lung-on-a-chip, HCC827 cells with CAFs co-culture or IL-6 treatment developed resistance to osimertinib. Further use of the IL-6 antibody inhibitor tocilizumab could reverse EGFR-TKI resistance to a certain extent. Combination therapy with tocilizumab and EGFR-TKIs may provide a novel therapeutic strategy for overcoming EGFR-TKI resistance caused by EMT in NSCLC. Furthermore, the lung-on-a-chip can simulate complex TME and can be used for evaluating tumour resistance and exploring mechanisms, with the potential to become an important tool for personalised diagnosis, treatment, and biomedical research.

2.
Mol Ther ; 32(5): 1561-1577, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38454607

ABSTRACT

Inflammation resolution is an essential process for preventing the development of chronic inflammatory diseases. However, the mechanisms that regulate inflammation resolution in psoriasis are not well understood. Here, we report that ANKRD22 is an endogenous negative orchestrator of psoriasiform inflammation because ANKRD22-deficient mice are more susceptible to IMQ-induced psoriasiform inflammation. Mechanistically, ANKRD22 deficiency leads to excessive activation of the TNFRII-NIK-mediated noncanonical NF-κB signaling pathway, resulting in the hyperproduction of IL-23 in DCs. This is due to ANKRD22 being a negative feedback regulator for NIK because it physically binds to and assists in the degradation of accumulated NIK. Clinically, ANKRD22 is negatively associated with IL-23A expression and psoriasis severity. Of greater significance, subcutaneous administration of an AAV carrying ANKRD22-overexpression vector effectively hastens the resolution of psoriasiform skin inflammation. Our findings suggest ANKRD22, an endogenous supervisor of NIK, is responsible for inflammation resolution in psoriasis, and may be explored in the context of psoriasis therapy.


Subject(s)
Disease Models, Animal , Interleukin-23 , Psoriasis , Signal Transduction , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/therapy , Psoriasis/etiology , Psoriasis/immunology , Psoriasis/genetics , Psoriasis/chemically induced , Animals , Mice , Interleukin-23/metabolism , Interleukin-23/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Mice, Knockout , Skin/pathology , Skin/metabolism , NF-kappaB-Inducing Kinase , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , NF-kappa B/metabolism
3.
Front Bioeng Biotechnol ; 11: 1299033, 2023.
Article in English | MEDLINE | ID: mdl-38026900

ABSTRACT

Lung cancer has become the primary cause of cancer-related deaths because of its high recurrence rate, ability to metastasise easily, and propensity to develop drug resistance. The wide-ranging heterogeneity of lung cancer subtypes increases the complexity of developing effective therapeutic interventions. Therefore, personalised diagnostic and treatment strategies are required to guide clinical practice. The advent of innovative three-dimensional (3D) culture systems such as organoid and organ-on-a-chip models provides opportunities to address these challenges and revolutionise lung cancer research and drug evaluation. In this review, we introduce the advancements in lung-related 3D culture systems, with a particular focus on lung organoids and lung-on-a-chip, and their latest contributions to lung cancer research and drug evaluation. These developments include various aspects, from authentic simulations and mechanistic enquiries into lung cancer to assessing chemotherapeutic agents and targeted therapeutic interventions. The new 3D culture system can mimic the pathological and physiological microenvironment of the lung, enabling it to supplement or replace existing two-dimensional culture models and animal experimental models and realize the potential for personalised lung cancer treatment.

4.
Phytomedicine ; 120: 155039, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37672855

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a significant global health concern that can lead to depression in affected patients. Liquiritin apioside (LA) possesses anti-oxidative and anti-inflammatory properties. However, its anti-inflammatory mechanism in IBD has not been extensively studied. PURPOSE: This study elucidates the pivotal role of LA in alleviating inflammation by regulating gut metabiota-derived metabolites and evaluating its regulative effects on promoting a balance of Th17/Treg cells in colitis mice. METHODS: To evaluate the effect of LA on IBD,16S rRNA gene sequencing and UPLC-QTOF-MS analysis were used to identify the changes of intestinal bacteria and their metabolites. Cytokines levels were determined by ELISA and qPCR, while immune cell ratios were evaluated via flow cytometry. RESULTS: Our findings revealed that LA treatment ameliorated general states of DSS-induced colitis mice and their accompanying depressive behaviors. Moreover, LA restricted the expression of pro-inflammatory cytokines and revised the imbalanced Treg/Th17 differentiation, while promoting SCFAs production in inflamed colon tissues. Fecal microbiota transplantation from LA-fed mice also corrected the imbalanced Treg/Th17 differentiation, indicating that LA-mediated restoration of the colonic Treg/Th17 balance mainly depends on the changes in gut metabolites. CONCLUSION: These results provide scientific evidence explaining the apparent paradox of low bioavailability and high bioactivity in polyphenols, and suggesting that LA could be used as a potential dietary supplement for the prevention and improvement of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Animals , Mice , Depression/drug therapy , RNA, Ribosomal, 16S , T-Lymphocytes, Regulatory , Colitis/drug therapy , Inflammation , Cytokines
5.
Cell Rep ; 42(8): 112910, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37531255

ABSTRACT

Amino acid (aa) metabolism is closely correlated with the pathogenesis of psoriasis; however, details on aa transportation during this process are barely known. Here, we find that SLC38A5, a sodium-dependent neutral aa transporter that counter-transports protons, is markedly upregulated in the psoriatic skin of both human patients and mouse models. SLC38A5 deficiency significantly ameliorates the pathogenesis of psoriasis, indicating a pathogenic role of SLC38A5. Surprisingly, SLC38A5 is almost exclusively expressed in dendritic cells (DCs) when analyzing the psoriatic lesion and mainly locates on the lysosome. Mechanistically, SLC38A5 potentiates lysosomal acidification, which dictates the cleavage and activation of TLR7 with ensuing production of pro-inflammatory cytokines such as interleukin-23 (IL-23) and IL-1ß from DCs and eventually aggravates psoriatic inflammation. In summary, this work uncovers an auxiliary mechanism in driving lysosomal acidification, provides inspiring insights for DC biology and psoriasis etiology, and reveals SLC38A5 as a promising therapeutic target for treating psoriasis.


Subject(s)
Amino Acid Transport Systems, Neutral , Psoriasis , Animals , Mice , Humans , Dendritic Cells/metabolism , Skin/pathology , Psoriasis/pathology , Inflammation/pathology , Disease Models, Animal , Lysosomes/pathology , Hydrogen-Ion Concentration
6.
Front Med (Lausanne) ; 10: 1167742, 2023.
Article in English | MEDLINE | ID: mdl-37497274

ABSTRACT

Objective: The purpose of this study was to develop a comprehensive nomogram for the cancer-specific survival (CSS) of white patients with invasive melanoma at back, posterior arm, posterior neck, and posterior scalp (BANS) sites and to determine the validity of the nomogram by comparing it with the conventional American Joint Committee on Cancer (AJCC) staging system. Methods: This study analyzed the patients with invasive melanoma in the Surveillance, Epidemiology, and End Results (SEER) database. R software was used to randomly divide the patients into training and validation cohorts at a ratio of 7:3. Multivariable Cox regression was used to identify predictive variables. The new survival nomogram was compared with the AJCC prognosis model using the concordance index (C-index), area under the receiver operating characteristic (ROC) curve (AUC), net reclassification index (NRI), integrated discrimination index (IDI), calibration plotting, and decision-curve analysis (DCA). Results: A novel nomogram was established to determine the 3-, 5-, and 8-year CSS probabilities of patients with invasive melanoma. According to the nomogram, the Age at Diagnosis had the greatest influence on CSS in invasive melanoma, followed by Bone Metastasis, AJCC, Stage, Liver Metastasis, Histologic Subtype, Brain Metastasis, Ulceration, and Primary Site. The nomogram had a higher C-index than the AJCC staging system in both the training (0.850 versus 0.799) and validation (0.829 versus 0.783) cohorts. Calibration plotting demonstrated that the model had good calibration ability. The nomogram outperformed the AJCC staging system in terms of AUC, NRI, IDI, and DCA. Conclusion: This was the first study to develop and evaluate a comprehensive nomogram for the CSS of white patients with invasive melanoma at BANS sites using the SEER database. The novel nomogram can assist clinical staff in predicting the 3-, 5-, and 8-year CSS probabilities of patients with invasive melanoma more accurately than can the AJCC staging system.

7.
Cell Rep ; 42(7): 112684, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37355989

ABSTRACT

γδ T cells make key contributions to tissue physiology and immunosurveillance through two main functionally distinct subsets, γδ T1 and γδ T17. m6A methylation plays critical roles in controlling numerous aspects of mRNA metabolism that govern mRNA turnover, gene expression, and cellular functional specialization; however, its role in γδ T cells remains less well understood. Here, we find that m6A methylation controls the functional specification of γδ T17 vs. γδ T1 cells. Mechanistically, m6A methylation prevents the formation of endogenous double-stranded RNAs and promotes the degradation of Stat1 transcripts, which converge to prevent over-activation of STAT1 signaling and ensuing inhibition of γδ T17. Deleting Mettl3, the key enzyme in the m6A methyltransferases complex, in γδ T cells reduces interleukin-17 (IL-17) production and ameliorates γδ T17-mediated psoriasis. In summary, our work shows that METTL3-mediated m6A methylation orchestrates mRNA stability and double-stranded RNA (dsRNA) contents to equilibrate γδ T1 and γδ T17 cells.


Subject(s)
Methyltransferases , RNA, Double-Stranded , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
J Med Virol ; 95(2): e28475, 2023 02.
Article in English | MEDLINE | ID: mdl-36606607

ABSTRACT

Global coronavirus disease 2019 (COVID-19) pandemics highlight the need of developing vaccines with universal and durable protection against emerging SARS-CoV-2 variants. Here we developed an extended-release vaccine delivery system (GP-diABZI-RBD), consisting the original SARS-CoV-2 WA1 strain receptor-binding domain (RBD) as the antigen and diABZI stimulator of interferon genes (STING) agonist in conjunction with yeast ß-glucan particles (GP-diABZI) as the platform. GP-diABZI-RBD could activate STING pathway and inhibit SARS-CoV-2 replication. Compared to diABZI-RBD, intraperitoneal injection of GP-diABZI-RBD elicited robust cellular and humoral immune responses in mice. Using SARS-CoV-2 GFP/ΔN transcription and replication-competent virus-like particle system (trVLP), we demonstrated that GP-diABZI-RBD-prototype vaccine exhibited the strongest and durable humoral immune responses and antiviral protection; whereas GP-diABZI-RBD-Omicron displayed minimum neutralization responses against trVLP. By using pseudotype virus (PsVs) neutralization assay, we found that GP-diABZI-RBD-Prototype, GP-diABZI-RBD-Delta, and GP-diABZI-RBD-Gamma immunized mice sera could efficiently neutralize Delta and Gamma PsVs, but had weak protection against Omicron PsVs. In contrast, GP-diABZI-RBD-Omicron immunized mice sera displayed the strongest neutralization response to Omicron PsVs. Taken together, the results suggest that GP-diABZI can serve as a promising vaccine delivery system for enhancing durable humoral and cellular immunity against broad SARS-CoV-2 variants. Our study provides important scientific basis for developing SARS-CoV-2 VOC-specific vaccines.


Subject(s)
COVID-19 , Vaccines , Animals , Humans , Mice , SARS-CoV-2 , COVID-19 Vaccines , Immunity, Cellular , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus , Antibodies, Viral
9.
Appl Opt ; 61(31): 9124-9134, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36607045

ABSTRACT

Image encryption has emerged as a method of disguising an image with a noisy or meaningless appearance to prevent its content from being accessed by unauthorized users. We propose an architecture named flexible image encryption and decryption ResNet (FEDResNet) for diffusing an image in end-to-end mode. The architecture consists of an encryption network for diffusing the image and a decryption network for restoring the plaintext image from the diffused image. To enhance the security of the encrypted image, the diffused image is further processed with two optional operations: parallel scrambling and serial diffusion. Two key planes are constructed based on a user-defined key with a chaotic map to control the authority to access images. The structure and parameters of FEDResNet can be shared publicly by different users; hence, it is more flexible and convenient than previous deep-learning-based image encryption methods. A classification network is trained to classify medical images in ciphertext environments. The proposed FEDResNet is trained and tested on the ImageNet data set. Extensive experiments have been performed, and the experimental results suggest that the proposed model can achieve a high level of security with satisfactory efficiency. The experimental results also show that FEDResNet-encrypted images can be classified directly in the ciphertext domain by authorized users as accurately as plaintext images, which is a superior property that is not possessed by traditional image encryption methods.

10.
Cancer Lett ; 526: 322-334, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34767926

ABSTRACT

The relationship between microRNA (miRNA) and hosting long non-coding RNA (lncRNA) remains unclear. Here, the expression levels of microRNA-210 (miR-210) and hosting lncRNA MIR210HG are significantly increased and positively correlated in gastric cancer (GC). Gain- and loss-of-function studies demonstrate that miR-210 and MIR210HG synergistically promote the migration and invasion of GC cells in vitro. Furthermore, GC sublines simultaneously expressing miR-210 and MIR210HG display synergistic promotion of lung metastasis in vivo. Mechanistically, MIR210HG interacts with DExH-box helicase 9 (DHX9) to increase DHX9/c-Jun complex's occupancy on the promoter of matrix metallopeptidases (MMPs), and thus promotes migration and invasion of GC cells. Additionally, miR-210 directly suppresses the expression of dopamine receptor D5 (DRD5), serine/threonine kinase 24 (STK24) and MAX network transcriptional repressor (MNT), resulting in enhanced migration and invasion. Finally, MYC proto-oncogene (c-Myc) transactivates miR-210 and MIR210HG. Overexpression of miR-210 or/and MIR210HG can rescue the inhibitory effect on the migration and invasion by silencing c-Myc. Moreover, c-Myc inhibitor significantly decreases lung metastasis of GC in vivo. Collectively, our findings identify a novel mechanism, by which c-Myc-activated miR-210 and MIR210HG synergistically promote the metastasis of GC.


Subject(s)
MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Animals , Cell Line, Tumor , Female , Genes, myc , Heterografts , Humans , Introns , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplasm Metastasis , Proto-Oncogene Proteins c-myc/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
11.
Nature ; 600(7888): 314-318, 2021 12.
Article in English | MEDLINE | ID: mdl-34819664

ABSTRACT

Thermogenesis in brown and beige adipose tissue has important roles in maintaining body temperature and countering the development of metabolic disorders such as obesity and type 2 diabetes1,2. Although much is known about commitment and activation of brown and beige adipose tissue, its multiple and abundant immunological factors have not been well characterized3-6. Here we define a critical role of IL-27-IL-27Rα signalling in improving thermogenesis, protecting against diet-induced obesity and ameliorating insulin resistance. Mechanistic studies demonstrate that IL-27 directly targets adipocytes, activating p38 MAPK-PGC-1α signalling and stimulating the production of UCP1. Notably, therapeutic administration of IL-27 ameliorated metabolic morbidities in well-established mouse models of obesity. Consistently, individuals with obesity show significantly decreased levels of serum IL-27, which can be restored after bariatric surgery. Collectively, these findings show that IL-27 has an important role in orchestrating metabolic programs, and is a highly promising target for anti-obesity immunotherapy.


Subject(s)
Adipocytes/metabolism , Energy Metabolism , Interleukin-27/metabolism , Thermogenesis , Animals , Bariatric Surgery , Disease Models, Animal , Female , Humans , Insulin Resistance , Interleukin-27/blood , Interleukin-27/therapeutic use , Male , Mice , Obesity/blood , Obesity/drug therapy , Obesity/metabolism , Obesity/prevention & control , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Uncoupling Protein 1/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
12.
ACS Appl Mater Interfaces ; 13(34): 40415-40428, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34470103

ABSTRACT

Toxoplasma gondii (T. gondii) infection causes severe zoonotic toxoplasmosis, which threatens the safety of almost one-third of the human population globally. However, there is no effective protective vaccine against human toxoplasmosis. This necessitates anti-T. gondii vaccine development, which is a main priority of public health. In this study, we optimized the adjuvant system 04 (AS04), a vaccine adjuvant constituted by 3-O-desacyl-4'-monophosphoryl lipid A (a TLR4 agonist) and aluminum salts, by packing it within natural extracts of ß-glucan particles (GPs) from Saccharomyces cerevisiae to form a GP-AS04 hybrid adjuvant system. Through a simple mixing procedure, we loaded GP-AS04 particles with the total extract (TE) of T. gondii lysate, forming a novel anti-T. gondii vaccine GP-AS04-TE. Results indicated that the hybrid adjuvant can efficiently and stably load antigens, mediate antigen delivery, facilitate the dendritic uptake of antigens, boost dendritic cell maturation and stimulation, and increase the secretion of pro-inflammatory cytokines. In the mouse inoculation model, GP-AS04-TE significantly stimulated the function of dendritic cells, induced a very strong TE-specific humoral and cellular immune response, and finally showed a strong and effective protection against toxoplasma chronic and acute infections. This work proves the potential of GP-AS04 for exploitation as a vaccine against a range of pathogens.


Subject(s)
Adjuvants, Vaccine/therapeutic use , Aluminum Hydroxide/therapeutic use , Lipid A/analogs & derivatives , Nanocomposites/therapeutic use , Protozoan Vaccines/therapeutic use , Toxoplasma/immunology , Toxoplasmosis/prevention & control , Adjuvants, Vaccine/chemistry , Adjuvants, Vaccine/toxicity , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/immunology , Aluminum Hydroxide/toxicity , Animals , Dendritic Cells/drug effects , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/therapeutic use , Fungal Polysaccharides/toxicity , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Lipid A/chemistry , Lipid A/immunology , Lipid A/therapeutic use , Lipid A/toxicity , Male , Mice, Inbred C57BL , Nanocomposites/chemistry , Nanocomposites/toxicity , Phagocytes/drug effects , Protozoan Vaccines/chemistry , Protozoan Vaccines/immunology , Protozoan Vaccines/toxicity , Saccharomyces cerevisiae/chemistry , Tissue Extracts/chemistry , Tissue Extracts/immunology , Tissue Extracts/therapeutic use , Tissue Extracts/toxicity , Toxoplasma/chemistry , Toxoplasmosis/immunology , beta-Glucans/chemistry , beta-Glucans/therapeutic use , beta-Glucans/toxicity
13.
J Clin Invest ; 130(10): 5180-5196, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32831293

ABSTRACT

Psoriasis is a severe disease associated with the disturbance of metabolism and inflammation, but the molecular mechanisms underlying these aspects of psoriasis pathology are poorly understood. Here, we report that glutaminase 1-mediated (GLS1-mediated) glutaminolysis was aberrantly activated in patients with psoriasis and in psoriasis-like mouse models, which promoted Th17 and γδ T17 (IL-17A-producing γδ T) cell differentiation through enhancement of histone H3 acetylation of the Il17a promoter, thereby contributing to the immune imbalance and development of psoriasis. We further demonstrate that mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) protease was constitutively active in psoriatic CD4+ and γδ T cells, thereby supporting GLS1 expression by stabilizing c-Jun, which directly binds to the GLS1 promoter region. Blocking the activity of either GLS1 or MALT1 protease resolved Th17 and γδ T17 cell differentiation and epidermal hyperplasia in the psoriasis-like mouse models. Finally, IL-17A enhanced GLS1 expression via the MALT1/cJun pathway in keratinocytes, resulting in hyperproliferation of and chemokine production by keratinocytes. Our findings identify the role of the MALT1/cJun/GLS1/glutaminolysis/H3 acetylation/T17 axis in psoriasis pathogenesis and reveal potential therapeutic targets for this disease.


Subject(s)
Glutaminase/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Psoriasis/etiology , Psoriasis/metabolism , Adult , Aged , Animals , Case-Control Studies , Cell Differentiation/immunology , Disease Models, Animal , Female , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutamine/metabolism , Humans , Interleukin-17/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Psoriasis/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult
14.
Front Immunol ; 11: 846, 2020.
Article in English | MEDLINE | ID: mdl-32528465

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the main causes of cirrhosis and major risk factors for hepatocellular carcinoma and liver-related death. Despite substantial clinical and basic research, the pathogenesis of obesity-related NAFLD remains poorly understood. In this study, we show that perforin can act as an immune regulator to prevent the progression of NAFLD. Aged perforin-deficient (Prf-/-) mice have increased lipid accumulation in the liver compared to WT mice. With high-fat diet (HFD) challenge, Prf-/- mice have increased liver weight, more severe liver damage, and increased liver inflammation when compared with WT controls. Mechanistic studies revealed that perforin specifically regulates intrinsic IFN-γ production in CD4 T cells, not CD8 T cells. We found that CD4 T cell depletion reduces liver injury and ameliorates the inflammation and metabolic morbidities in Prf-/- mice. Furthermore, improved liver characteristics in HFD Prf-/- and IFN-γR-/- double knockout mice confirmed that IFN-γ is a key factor for mediating perforin regulation of NAFLD progression. Overall, our findings reveal the important regulatory role perforin plays in the progression of obesity-related NAFLD and highlight novel strategies for treating NAFLD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Disease Progression , Interferon-gamma/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Perforin/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatitis/etiology , Interferon-gamma/genetics , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/blood , Obesity/metabolism , Perforin/deficiency , Perforin/genetics
15.
J Immunol Res ; 2020: 5084682, 2020.
Article in English | MEDLINE | ID: mdl-32352019

ABSTRACT

Type I interferon kappa (IFNκ) is selectively expressed in human keratinocytes. Herpes simplex virus-1 (HSV-1) is a human pathogen that infects keratinocytes and causes lytic skin lesions. Whether IFNκ plays a role in keratinocyte host defense against HSV-1 has not been investigated. In this study, we found that IFNκ mRNA expression was induced by addition of recombinant IFNκ and poly (I:C); and its expression level was significantly greater than IFNa2, IFNb1, and IFNL1 in both undifferentiated and differentiated normal human epidermal keratinocytes (NHEKs) under resting and stimulation conditions. Although IFNe was expressed at a relatively higher level than other IFNs in resting undifferentiated NHEK, its expression level did not change after stimulation with recombinant IFNκ and poly (I:C). HSV-1 infection inhibited gene expression of IFNκ and IFNe in NHEK. Silencing IFNκ in NHEK led to significantly enhanced HSV-1 replication in both undifferentiated and differentiated NHEK compared to scrambled siRNA-transfected cells, while the addition of recombinant IFNκ significantly reduced HSV-1 replication in NHEK. In addition, we found that IFNκ did not regulate protein expression of NHEK differentiation markers. Our results demonstrate that IFNκ is the dominant type of IFNs in keratinocytes and it has an important function for keratinocytes to combat HSV-1 infection.


Subject(s)
Herpes Simplex/immunology , Herpesvirus 1, Human/physiology , Interferon Type I/metabolism , Keratinocytes/physiology , RNA, Small Interfering/genetics , Skin/pathology , Cell Differentiation , Cell Line , Humans , Immunity, Innate , Interferon Type I/genetics , Poly I-C/immunology , Up-Regulation , Virus Replication
16.
Clin Cosmet Investig Dermatol ; 12: 865-873, 2019.
Article in English | MEDLINE | ID: mdl-31819584

ABSTRACT

PURPOSE: There is increased type I interferon signature in psoriasis patients. Interferon-kappa (IFN-κ) is a member of type I interferon family that is constitutively expressed by keratinocytes. In this study, we investigate whether IFN-κ is involved in psoriasis etiology. PATIENTS AND METHODS: Twenty healthy individuals, 20 psoriasis vulgaris patients and 10 atopic dermatitis (AD) were included for this study. Immunohistochemistry staining, normal human epidermal keratinocytes (NHEK) culture, Ca2Cl-induced differentiation, quantitative reverse transcription (qRT-PCR), ELISA and murine experiments were performed. RESULTS: We found IFN-κ protein expression was extremely low in the epidermis of normal skin, but it was significantly increased in the suprabasal layers of epidermal keratinocytes in psoriatic skin lesions. However, its expression in the skin lesions of AD was similar to normal skin. Additionally, IFN-κ protein was detected in sera from psoriasis patients, but not in sera from normal subjects and AD. We further investigated the regulation of IFNk gene expression in NHEK. We found that IFNk was significantly induced by types of nucleic acid pathogen recognition receptor (PRR) agonists in NHEK. While its expression was significantly induced by itself and IFN-γ, it was inhibited by type 2 immunity cytokines IL4 and IL13; other inflammatory cytokines including IL1 super-family members and IL17A did not alter its expression. Addition of recombinant IFN-κ did not affect keratinocytes differentiation. Using the murine experimental model, we demonstrated that subcutaneous administration of recombinant IFN-κ did not increase skin thickness, but significantly increased the transcription of TNFA and IL17A in mice skin. CONCLUSION: Increased IFN-κ in psoriasis may be caused by injured cells-released nucleic acids, increased IFN-γ and self-activation. Its enhancement may contribute to the etiology of the disease by enhancing TNFA and IL17A gene expression.

17.
Mol Carcinog ; 58(12): 2340-2352, 2019 12.
Article in English | MEDLINE | ID: mdl-31556968

ABSTRACT

Mucosa-associated lymphoid tissue lymphoma translocation protein-1 (MALT1) protease presents crucial antiapoptotic properties in activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL); however, the mechanism is unclear. Here, we reported that inhibition of MALT1 protease in ABC-DLBCL cells led to cell apoptosis, along with elevated mitochondrial reactive oxygen species production and a reduced oxygen consumption rate. These alterations induced by MALT1 protease inhibition were associated with reduced expression of glutaminase (GLS1) and glutathione levels. We further show that MALT1 protease was required for the activation and nuclear translocation of c-Jun, which functions as a transcription factor of the GLS1 gene by binding directly to its promoter region. Taken together, MALT1 protease maintained mitochondrial redox homeostasis and mitochondrial bioenergetics through the MALT1-c-Jun-GLS1-coupled metabolic pathway to defend against apoptosis in ABC-DLBCL cells, which raises exciting possibilities regarding targeting of the MALT1-c-Jun-GLS1 axis as a potential therapeutic strategy against ABC-DLBCL.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Glutaminase/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Proto-Oncogene Proteins c-jun/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Glutaminase/metabolism , Glutathione/metabolism , Homeostasis , Humans , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mitochondria/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins c-jun/metabolism
18.
J Agric Food Chem ; 67(34): 9522-9531, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31379161

ABSTRACT

The imbalance of T lymphocyte subsets substantially conduces to disturbed intestinal immune system and succeeding colonic tissue damage in inflammatory bowel diseases. It is considered that regulation of phytochemicals on cytokine production potentially provides a broad prospect for the exploitation of immunomodulatory agents. Here, we reported that oral administration of feruloylated oligosaccharides (FOs) effectively alleviated mice colitis disease induced by dextran sulfate sodium (DSS). FOs decreased the percentage of T helper (Th)17 cells and downregulated the production of Th17-specific cytokines. In contrast, FOs increased the percentage of regulatory T (Treg) cells and elevated the production of Treg-specific cytokines in colons of DSS-challenged mice. These results indicated that FOs restored the immunologic equilibrium of Th17 and Treg subsets, hereby ameliorating the deterioration of colitis. Furthermore, FOs diminished the secretion of interleukin (IL)-23 and IL-6 but enhanced the transforming growth factor-ß1 (TGF-ß1) in dendritic cells in vitro and in vivo, which contributed to the restoration of Th17 and Treg cells immune balance. The mechanistic analysis showed that the regulation of FOs on IL-23 and IL-6 was associated with the nuclear factor-κ-gene binding signaling pathway and TGF-ß1 with mitogen-activated protein kinase-activator protein 1 signaling pathway. Taken together, oral administration of FOs exerted potent immunomodulatory effects against mice colitis via restoring the immune balance of Th17 and Treg cells.


Subject(s)
Colitis/drug therapy , Oligosaccharides/administration & dosage , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Cytokines/genetics , Cytokines/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Interleukin-23/genetics , Interleukin-23/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Mice, Inbred C57BL , Oligosaccharides/chemistry , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
19.
J Immunol Res ; 2018: 5214187, 2018.
Article in English | MEDLINE | ID: mdl-30069489

ABSTRACT

The innate immune response is the first line defense against viral infections. Novel genes involved in this system are continuing to emerge. SLC15A3, a proton-coupled histidine and di-tripeptide transporter that was previously found in lysosomes, has been reported to inhibit chikungunya viral replication in host cells. In this study, we found that SLC15A3 was significantly induced by DNA virus herpes simplex virus-1(HSV-1) in monocytes from human peripheral blood mononuclear cells. Aside from monocytes, it can also be induced by HSV-1 in 293T, HeLa cells, and HaCaT cells. Overexpression of SLC15A3 in 293T cells inhibits HSV-1 replication and enhances type I and type III interferon (IFN) responses, while silencing SLC15A3 leads to enhanced HSV-1 replication with reduced IFN production. Moreover, we found that SLC15A3 interacted with MAVS and STING and potentiated MAVS- and STING-mediated IFN production. These results demonstrate that SLC15A3 participates in anti-HSV-1 innate immune responses by regulating MAVS- and STING-mediated signaling pathways.


Subject(s)
Herpes Simplex/immunology , Immunity, Innate/immunology , Membrane Transport Proteins/immunology , Adaptor Proteins, Signal Transducing/immunology , Herpesvirus 1, Human , Humans , Membrane Proteins/immunology , Signal Transduction/immunology
20.
PLoS One ; 13(1): e0191127, 2018.
Article in English | MEDLINE | ID: mdl-29304137

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0167392.].

SELECTION OF CITATIONS
SEARCH DETAIL
...