Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 17495, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39079980

ABSTRACT

Short-chain fatty acids (SCFAs) have been proposed to have anti-inflammatory effects and improve immune homeostasis. We aimed to examine the effects of SCFAs on skin phenotype, systemic inflammation, and gut microbiota in mice with psoriasis-like inflammation. Imiquimod (IMQ)-treated C57BL/6 mice served as the study model. We conducted a metagenomic association study of IMQ-mice treated with SCFAs or anti-IL-17 antibody using whole-genome shotgun sequencing. The associations among SCFA supplements, skin thickness, circulating inflammatory profiles, and fecal microbiota profiles were investigated. The microbiome study was performed using pipelines for phylogenetic analysis, functional gene analysis, and pathway analysis. In IMQ-treated mice, there were increases in skin thickness and splenic weight, as well as unique fecal microbial profiles. SCFAs ameliorated IMQ-induced skin thickening, splenic weight gain, and serum IL-17F levels, with results that were comparable with those receiving anti-IL-17 treatment. IMQ-treated mice receiving SCFAs had greater microbial diversity than mice treated with IMQ alone. SCFAs and anti-IL17 treatment were associated with alteration of gut microbiota, with increased prevalences of Oscillospiraceae and Lachnopiraceae and decreased prevalences of Muribaculaceae and Bacteroides, which have been predicted to be associated with increased glycan degradation, phenylalanine metabolism, and xylene degradation. SCFAs may mitigate IMQ-induced skin thickening and IL-17F levels and alter fecal microbiota profiles in IMQ-treated mice.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Imiquimod , Interleukin-17 , Mice, Inbred C57BL , Skin , Animals , Imiquimod/adverse effects , Gastrointestinal Microbiome/drug effects , Interleukin-17/metabolism , Fatty Acids, Volatile/metabolism , Mice , Skin/drug effects , Skin/pathology , Skin/microbiology , Skin/metabolism , Metagenomics/methods , Psoriasis/drug therapy , Psoriasis/chemically induced , Psoriasis/microbiology , Metagenome , Feces/microbiology
2.
Adv Sci (Weinh) ; 10(17): e2206856, 2023 06.
Article in English | MEDLINE | ID: mdl-37078788

ABSTRACT

HLA-G is considered as an immune checkpoint protein and a tumor-associated antigen. In the previous work, it is reported that CAR-NK targeting of HLA-G can be used to treat certain solid tumors. However, the frequent co-expression of PD-L1 and HLA-G) and up-regulation of PD-L1 after adoptive immunotherapy may decrease the effectiveness of HLA-G-CAR. Therefore, simultaneous targeting of HLA-G and PD-L1 by multi-specific CAR could represent an appropriate solution. Furthermore, gamma-delta T (γδT) cells exhibit MHC-independent cytotoxicity against tumor cells and possess allogeneic potential. The utilization of nanobodies offers flexibility for CAR engineering and the ability to recognize novel epitopes. In this study, Vδ2 γδT cells are used as effector cells and electroporated with an mRNA-driven, nanobody-based HLA-G-CAR with a secreted PD-L1/CD3ε Bispecific T-cell engager (BiTE) construct (Nb-CAR.BiTE). Both in vivo and in vitro experiments reveal that the Nb-CAR.BiTE-γδT cells could effectively eliminate PD-L1 and/or HLA-G-positive solid tumors. The secreted PD-L1/CD3ε Nb-BiTE can not only redirect Nb-CAR-γδT but also recruit un-transduced bystander T cells against tumor cells expressing PD-L1, thereby enhancing the activity of Nb-CAR-γδT therapy. Furthermore, evidence is provided that Nb-CAR.BiTE redirectes γδT into tumor-implanted tissues and that the secreted Nb-BiTE is restricted to the tumor site without apparent toxicity.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , B7-H1 Antigen/metabolism , HLA-G Antigens/metabolism , Receptors, Chimeric Antigen/metabolism
3.
J Invest Dermatol ; 141(9): 2219-2228.e8, 2021 09.
Article in English | MEDLINE | ID: mdl-33744296

ABSTRACT

Lysosomal adaptation is a cellular physiological process in which the number and function of lysosomes are regulated at the transcriptional and post-transcriptional levels in response to extracellular and/or intracellular cues or lysosomal damage. Imiquimod (IMQ), a synthetic toll-like receptor 7 ligand with hydrophobic and weak basic properties, exhibits both antitumor and antiviral activity against various skin malignancies as a clinical treatment. Interestingly, IMQ has been suggested to be highly concentrated in the lysosomes of plasmacytoid dendritic cells, indicating that IMQ could modulate lysosome function after sequestration in the lysosome. In this study, we found that IMQ not only induced lysosomal membrane permeabilization and dysfunction but also increased lysosome biogenesis to achieve lysosomal adaptation in cancer cells. IMQ-induced ROS production but not lysosomal sequestration of IMQ was the major cause of lysosomal adaptation. Moreover, IMQ-induced lysosomal adaptation occurred through lysosomal calcium ion release and activation of the calcineurin/TFEB axis to promote lysosome biogenesis. Finally, depletion of TFEB sensitized skin cancer cells to IMQ-induced apoptosis in vitro and in vivo. In summary, a disruption of lysosomal adaptation might represent a therapeutic strategy for synergistically enhancing the cytotoxicity of IMQ in skin cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Imiquimod/therapeutic use , Lysosomes/drug effects , Stomach Neoplasms/drug therapy , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Calcineurin/metabolism , Calcium Signaling , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
4.
J Dermatol Sci ; 98(3): 152-162, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32376151

ABSTRACT

BACKGROUND: Mitochondrial homeostasis is a highly dynamic process involving continuous fission and fusion cycles and mitophagy to maintain mitochondrial functionality. Imiquimod (IMQ), a Toll-like receptor (TLR) 7 ligand, is used to treat various skin malignancies. IMQ also induces apoptotic and autophagic cell death in various cancers through a TLR7-independent pathway. OBJECTIVE: To investigate whether IMQ-induced ROS production is involved in mitochondrial dysfunction, mitochondrial fragmentation and mitophagy in skin cancer cells. METHODS: BCC/KMC-1, B16F10 and A375 skin cancer cells, AGS gastric cancer cells and primary human keratinocytes were treated with 50 µg/mL IMQ. After 4 h, ROS were detected by CM-H2DCFDA, DHE, and MitoSOX Red staining. After 24 h, cell viability and the mitochondrial membrane potential were evaluated by a CCK-8 assay and JC-1 staining, respectively. Oxygen consumption was assessed with an Oroboros instrument. Mitochondrial morphology and mitophagy were evaluated by MitoTracker and LysoTracker staining. Mitochondrial dynamics markers, including MFN-1, DRP-1 and OPA1, and mitophagy markers, including LC3, S65-phosphorylated ubiquitin, PINK1 and TOM20, were detected by immunoblotting. RESULTS: IMQ not only induced severe ROS production but also resulted in increased mitochondrial membrane potential loss, mitochondrial fission and mitophagy and decreased oxygen consumption in skin cancer cells compared with normal keratinocytes. Pretreatment with the antioxidant NAC reduced IMQ-induced ROS production and attenuated IMQ-induced mitochondrial fission and mitophagy in skin cancer cells. CONCLUSIONS: IMQ-induced ROS might be associated with mitochondrial dysfunction, mitochondrial fission and mitophagy in cancer cells. Alleviating IMQ-induced ROS production would reduce mitochondrial fission-to-fusion skewing and further reduce IMQ-induced mitophagy.


Subject(s)
Antineoplastic Agents/pharmacology , Imiquimod/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Antioxidants/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Free Radical Scavengers/pharmacology , Humans , Imiquimod/therapeutic use , Keratinocytes , Mice , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Signal Transduction/drug effects , Skin Neoplasms/pathology
5.
Cell Death Discov ; 6: 17, 2020.
Article in English | MEDLINE | ID: mdl-32257389

ABSTRACT

Hepatocellular carcinoma (HCC), a hepatic malignancy, has a poor prognosis and contributes to cancer-related death worldwide. Cellular senescence is an anticancer therapeutic strategy that causes irreversible cell cycle arrest and enables immune-mediated clearance of cancer cells. Atorvastatin, an HMG-CoA reductase inhibitor, has been shown to inhibit tumor growth and induce apoptosis or autophagy in malignant tumors. However, whether atorvastatin can induce HCC cell senescence and the mechanisms involved are poorly understood. The effects of atorvastatin-induced senescence were examined in both HCC cells and mouse xenograft models. The phenomenon and mechanism of senescence were examined by cell cycle analysis, senescence-associated ß-galactosidase (SA-ß-gal) staining and western blotting in HCC cells, and HCC tissues from mice were analyzed by immunohistochemical (IHC) staining. We demonstrated that atorvastatin induced cell growth inhibition and G0/G1 phase cell cycle arrest, leading to senescence in HCC cells. Atorvastatin-induced senescence was independent of p53, p14, and p16, and atorvastatin not only decreased the secretion of IL-6, a major senescence-associated secretory phenotype (SASP) factor, and the phosphorylation of STAT3 but also inhibited the expression of hTERT, a catalytic subunit of telomerase. Supplementation with exogenous IL-6 reversed both atorvastatin-induced suppression of STAT3 phosphorylation and hTERT expression and atorvastatin-induced senescence. Overexpression of constitutively activated STAT3 rescued HCC cells from atorvastatin-induced hTERT suppression and senescence. Moreover, atorvastatin decreased tumor growth in mouse xenograft models. Consistent with these results, atorvastatin decreased the IL-6, p-STAT3, and hTERT levels and increased ß-gal expression in tumor sections. Taken together, these data indicate that atorvastatin can induce atypical cellular senescence in HCC cells to inhibit tumor growth, an effect mediated by downregulation of hTERT through suppression of the IL-6/STAT3 pathway.

6.
J Invest Dermatol ; 140(9): 1771-1783.e6, 2020 09.
Article in English | MEDLINE | ID: mdl-32035924

ABSTRACT

The induction of immunogenic cell death (ICD) in cancer cells triggers specific immune responses against the same cancer cells. Imiquimod (IMQ) is a synthetic ligand of toll-like receptor 7 that exerts antitumor activity by stimulating cell-mediated immunity or by directly inducing apoptosis. Whether IMQ causes tumors to undergo ICD and elicits a specific antitumor immune response is unknown. We demonstrated that IMQ-induced ICD-associated features, including the surface exposure of calreticulin and the secretion of adenosine triphosphate and HMGB1, were mediated by ROS and endoplasmic reticulum stress. In a B16F10 melanoma mouse model, vaccinating mice with IMQ-induced ICD cell lysate or directly injecting IMQ in situ reduced tumor growth that was mediated by inducing tumor-specific T-cell proliferation, promoting tumor-specific cytotoxic killing by CD8+ T cells, and increasing the infiltration of various immune cells into tumor lesions. The ICD-associated features were crucial in the induction of specific antitumor immunity in vivo. The glycolytic inhibitor 2-deoxyglucose enhanced IMQ-induced ICD-associated features and strengthened the antitumor immunity mediated by IMQ-induced ICD cell lysate in p53-mutant cancer cells, which were IMQ-resistant in vitro. We conclude that IMQ is an authentic ICD inducer and provide a concept connecting IMQ-induced cancer cell death and antitumor immune responses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxyglucose/pharmacology , Imiquimod/pharmacology , Immunogenic Cell Death/drug effects , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/transplantation , Deoxyglucose/therapeutic use , Drug Synergism , Glycolysis/drug effects , Humans , Imiquimod/therapeutic use , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Skin Neoplasms/immunology , Skin Neoplasms/pathology
7.
J Dermatol Sci ; 87(2): 138-148, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28601430

ABSTRACT

BACKGROUND: Autophagy is a highly conserved cellular catabolic pathway for degradation and recycling of intracellular components in response to nutrient starvation or environmental stress. Endoplasmic reticulum (ER) homeostasis can be disturbed by physiological and pathological influences, resulting in accumulation of misfolded and unfolded proteins in the ER lumen, a condition referred to as ER stress. Imiquimod (IMQ), a Toll-like receptor (TLR) 7 ligand, possesses anti-tumor and anti-viral activities in vitro and in vivo. OBJECTIVE: IMQ has been reported to promote the apoptosis of THP-1-derived macrophages through an ER stress-dependent pathway. However, the role of ER stress in IMQ-induced autophagy is unknown. In this study, we investigated the relationship between ER stress and IMQ-induced autophagy. METHODS: The expression of LC3, P62, p-PERK, Grp78, p-elF2α and IRE1α proteins were determined by immunoblotting. The relationship between ER stress and IMQ-induced autophagy were analyzed by ER stress inhibitors, a PERK inhibitor and the genetic silencing of PERK. The role of double-strand RNA-dependent protein kinase (PKR) activation in IMQ-induced autophagy was assessed by inhibiting PKR and genetically silencing PKR. The IMQ-induced autophagy was evaluated by immunoblotting and EGFP-LC3 puncta formation. RESULTS: IMQ induced reactive oxygen species (ROS) production in cancer cells. Additionally, IMQ markedly induced ER stress via ROS production and increased autophagosome formation in a dose- and time-dependent manner in both TLR7/8-expressing and TLR7/8-deficient cancer cells. Pharmacological or genetic inhibition of ER stress dramatically reduced LC3-II expression and EGFP-LC3 puncta formation in IMQ-treated cancer cells. IMQ-induced autophagy was markedly reduced by depletion and/or inhibition of PKR, a downstream effector of ER stress. CONCLUSION: IMQ-induced autophagy is dependent on PKR activation, which is mediated by ROS-triggered ER stress. These findings might provide useful information for basic research and for the clinical application of IMQ.


Subject(s)
Aminoquinolines/pharmacology , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , eIF-2 Kinase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , Fluorescent Antibody Technique , Heat-Shock Proteins/metabolism , Humans , Imiquimod , Indoles/pharmacology , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Toll-Like Receptor 7/metabolism , eIF-2 Kinase/genetics
8.
Cell Death Dis ; 8(2): e2626, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28230855

ABSTRACT

Hepatocellular carcinoma (HCC) is characterized by a poor prognosis and is one of the leading causes of cancer-related death worldwide. Simvastatin, an HMG-CoA reductase inhibitor, which decreases cholesterol synthesis by inhibiting mevalonate pathways and is widely used to treat cardiovascular diseases. Simvastatin exhibits anticancer effects against several malignancies. However, the molecular mechanisms underlying the anticancer effects of simvastatin on HCC are still not well understood. In this study, we demonstrated simvastatin-induced G0/G1 arrest by inducing p21 and p27 accumulation in HepG2 and Hep3B cells. Simvastatin also promoted AMP-activated protein kinase (AMPK) activation, which induced p21 upregulation by increasing its transcription. Consistent with this finding, we found genetic silencing of AMPK reduced p21 expression; however, AMPK silencing had no effect on p27 expression in HCC cells. Simvastatin decreased Skp2 expression at the transcriptional level, which resulted in p27 accumulation by preventing proteasomal degradation, an effect mediated by signal transducer and activator of transcription 3 (STAT3) inhibition. Constitutive STAT3 activation maintained high-level Skp2 expression and lower level p27 expression and significantly prevented G0/G1 arrest in simvastatin-treated HCC cells. Mevalonate decreased simvastatin-induced AMPK activation and rescued phospho-STAT3 and Skp2 expression in HCC cells, which resulted in the prevention of G0/G1 arrest through inhibition of p21 and p27 accumulation. Moreover, simvastatin significantly decreased tumor growth in HepG2 xenograft mice. Consistently, we found that simvastatin also increased p21 and p27 expression in tumor sections by reducing Skp2 expression and inducing AMPK activation and STAT3 suppression in the same tumor tissues. Taken together, these findings are demonstrative of the existence of a novel pathway in which simvastatin induces G0/G1 arrest by upregulating p21 and p27 by activating AMPK and inhibiting the STAT3-Skp2 axis, respectively. The results identify novel targets that explain the beneficial anticancer effects of simvastatin treatment on HCC in vitro and in vivo.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Liver Neoplasms/drug therapy , S-Phase Kinase-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Simvastatin/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase/drug effects , HEK293 Cells , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Resting Phase, Cell Cycle/drug effects , Signal Transduction/drug effects
9.
J Dermatol Sci ; 84(1): 59-70, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27449383

ABSTRACT

BACKGROUND: The activation of Toll-like receptor 7 (TLR7) in dendritic cells (DCs) plays a crucial role in the pathogenesis of psoriasis. The macrolide antibiotic azithromycin (AZM) had been demonstrated to inhibit the TLR4 agonist-induced maturation and activation of murine bone marrow-derived DCs (BMDCs). OBJECTIVE: To investigate the effects of AZM on the induction of DC maturation and activation by imiquimod (IMQ), a synthetic TLR7 agonist, as well as its potential as a therapeutic agent for psoriasis. METHODS: The effects of AZM on IMQ-induced DC activation were investigated based on the expression of cell surface markers and cytokine secretion. The lysosomal pH, post-translational processing of TLR7, and TLR7 signaling were also examined in DCs. The therapeutic effects of AZM on psoriasis were evaluated in a murine model of IMQ-induced psoriasis-like skin inflammation. RESULTS: AZM significantly inhibited the expression of co-stimulatory molecules (CD40 and CD80) and reduced TNF-α, IL-10, IL-12p40, IL-12p70, IL-23p19 in BMDCs and IFN-α production in plasmacytoid DCs. AZM treatment impaired lysosomal acidification, interrupted TLR7 maturation in the lysosome, and ultimately blocked the IMQ-induced NF-κB and IRF-7 nuclear translocation in DCs. AZM treatment decreased signs of IMQ-induced skin inflammation in BALB/c mice. In addition to decreasing keratinocyte hyper-proliferation and restoring their terminal differentiation, AZM treatment decreased the accumulation of DCs as well as CD4, CD8 T cells and IL-17 producing cells in psoriatic skin lesions. AZM treatment improved splenomegaly, decreased the populations of Th17 and γδ T cells, and reduced the expression of cytokines known to be involved in the pathogenesis of psoriasis, such as IL-17A, IL-17F, IL-22 and IL-23, in the skin and spleen. CONCLUSION: AZM impaired IMQ-induced DC activation by decreasing lysosomal acidification and disrupting TLR7 maturation and signaling. AZM significantly improved the IMQ-induced psoriasis-like inflammation in mice. AZM may be a potential therapeutic candidate for psoriasis treatment.


Subject(s)
Azithromycin/pharmacology , Dendritic Cells/cytology , Membrane Glycoproteins/metabolism , Psoriasis/immunology , Psoriasis/metabolism , Skin/metabolism , Toll-Like Receptor 7/metabolism , Aminoquinolines , Animals , Anti-Bacterial Agents/chemistry , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Imiquimod , Keratinocytes/cytology , Leukocytes, Mononuclear/cytology , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Signal Transduction , Skin/pathology , Spleen/metabolism , Th17 Cells/cytology
10.
J Dermatol Sci ; 81(3): 182-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26775629

ABSTRACT

BACKGROUND: The tumor suppressor p53 controls DNA repair, cell cycle, apoptosis, autophagy and numerous other cellular processes. Imiquimod (IMQ), a synthetic toll-like receptor (TLR) 7 ligand for the treatment of superficial basal cell carcinoma (BCC), eliminates cancer cells by activating cell-mediated immunity and directly inducing apoptosis and autophagy in cancer cells. OBJECTIVE: To evaluate the role of p53 in IMQ-induced cell death in skin cancer cells. METHODS: The expression, phosphorylation and subcellular localization of p53 were detected by real-time PCR, luciferase reporter assay, cycloheximide chase analysis, immunoblotting and immunocytochemistry. Using BCC/KMC1 cell line as a model, the upstream signaling of p53 activation was dissected by over-expression of TLR7/8, the addition of ROS scavenger, ATM/ATR inhibitors and pan-caspase inhibitor. The role of p53 in IMQ-induced apoptosis and autophagy was assessed by genetically silencing p53 and evaluated by a DNA content assay, immunoblotting, LC3 puncta detection and acridine orange staining. RESULTS: IMQ induced p53 mRNA expression and protein accumulation, increased Ser15 phosphorylation, promoted nuclear translocation and up-regulated its target genes in skin cancer cells in a TLR7/8-independent manner. In BCC/KMC1 cells, the induction of p53 by IMQ was achieved through increased ROS production to stimulate the ATM/ATR-Chk1/Chk2 axis but was not mediated by inducing DNA damage. The pharmacological inhibition of ATM/ATR significantly suppressed IMQ-induced p53 activation and apoptosis. Silencing of p53 significantly decreased the IMQ-induced caspase cascade activation and apoptosis but enhanced autophagy. Mutant p53 skin cancer cell lines were more resistant to IMQ-induced apoptosis than wildtype p53 skin cancer cell lines. CONCLUSION: IMQ induced ROS production to stimulate ATM/ATR pathways and contributed to p53-dependent apoptosis in a skin basal cell carcinoma cell line BCC/KMC1.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Basal Cell/drug therapy , Skin Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Autophagy/drug effects , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Imiquimod , Mutation , Phosphorylation , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics
11.
J Dermatol Sci ; 78(2): 108-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25766763

ABSTRACT

BACKGROUND: AMP-activated protein kinase (AMPK), a principal intracellular energy sensor, plays a crucial role in cell growth, proliferation, apoptosis and autophagy. Imiquimod (IMQ) directly exhibits anti-tumor activity through the induction of apoptosis and autophagic cell death. OBJECTIVE: To evaluate the role of AMPK in IMQ-induced apoptosis and autophagy. METHODS: The phosphorylation of AMPK and its substrates was detected by immunoblotting. ATP contents were analyzed by an ATP bioluminescence assay. The upstream signaling for AMPK activation was dissected by examination of TLR7/8 expression, over-expression of TLR7/8, the addition of AMPK kinase inhibitors, and the genetic silencing of Myd88 and LKB1. The role of AMPK activation in IMQ-induced autophagy and apoptosis was assessed by inhibiting AMPK, genetically silencing AMPK and over-expressing AMPK dominant-negative mutants. Autophagy and apoptosis were evaluated by a DNA content assay, immunoblotting, EGFP-LC3 puncta detection and acridine orange staining. RESULTS: IMQ could activate AMPK and autophagy in cancer cells not expressing TLR7/8. IMQ caused ATP depletion and induced LKB1-mediated AMPK activation. The down-regulation of AMPK activity via pharmacological inhibition and genetic silencing resulted in reduced IMQ-induced apoptosis but did not influence autophagy, and this rescue effect was associated with the retention of translation factor activity and anti-apoptotic Bcl-2 family member Mcl-1 protein expression levels. CONCLUSION: IMQ induces AMPK activation independent of TLR7/8 expression, resulting in translation inhibition and subsequent apoptosis through ATP depletion and LKB1 signaling, in skin tumor cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Basal Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Melanoma/metabolism , Protein Biosynthesis/drug effects , Skin Neoplasms/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Basal Cell/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Down-Regulation , Enzyme Activation/drug effects , Gene Silencing , Humans , Imiquimod , Melanoma/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism
12.
Oncotarget ; 5(5): 1363-81, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24658058

ABSTRACT

Tumor cells rely on aerobic glycolysis to maintain unconstrained cell growth and proliferation. Imiquimod (IMQ), a synthetic Toll-like receptor (TLR) 7/8 ligand, exerts anti-tumor effects directly by inducing cell death in cancer cells and/or indirectly by activating cellular immune responses against tumor cells. However, whether IMQ modulates glucose metabolism pathways remains unclear. In this study, we demonstrated that IMQ can enhance aerobic glycolysis by up-regulating HIF-1α expression at the transcriptional and translational levels via ROS mediated STAT3- and Akt-dependent pathways, independent of TLR7/8 signaling. The genetic silencing of HIF-1α not only repressed IMQ-induced aerobic glycolysis but also sensitized cells to IMQ-induced apoptosis due to faster ATP and Mcl-1 depletion. Moreover, the glucose analog 2-DG and the Hsp90 inhibitor 17-AAG, which destabilizes the HIF-1α protein, synergized with IMQ to induce tumor cell apoptosis in vitro and significantly inhibited tumor growth in vivo. Thus, we hypothesize that the IMQ-induced up-regulation of HIF-1α and aerobic glycolysis is a protective response to the metabolic stress generated by IMQ treatment, and thus, co-treatment with inhibitors of HIF-1α and/or glycolysis may be a useful therapeutic strategy to enhance the anti-tumor effects of IMQ in clinical settings.


Subject(s)
Aminoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Melanoma/drug therapy , Aminoquinolines/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Cell Line, Tumor , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Drug Therapy, Combination , Gene Silencing , Humans , Imiquimod , Keratinocytes , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Mice , Protein Biosynthesis/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics , Transcription, Genetic/drug effects , Up-Regulation/drug effects
13.
Cancer Immunol Immunother ; 59(8): 1259-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20390417

ABSTRACT

It is well known that DNA vaccines induce protective humoral and cell-mediated immune responses in several animal models. Antrodia camphorata (AC) is a unique basidiomycete fungus of the Polyporaceae family that only grows on the aromatic tree Cinnamomum kanehirai Hayata (Lauraceae) endemic to Taiwan. Importantly, AC has been shown to be highly beneficial in the treatment and prevention of cancer. The goal of this study was to investigate whether AC is able to augment the antitumor immune properties of a HER-2/neu DNA vaccine in a mouse model in which p185neu is overexpressed in MBT-2 tumor cells. Compared with the mice that received the HER-2/neu DNA vaccine alone, co-treatment with AC suppressed tumor growth and extended the survival rate. This increase in the antitumor efficacy was attributed to the enhancement of the Th1-like cellular immune response by the HER-2/neu DNA vaccine-AC combination. Evidence for this came from the marked increase in the IFN-gamma mRNA expression in CD4+ T cells in the draining inguinal lymph nodes, an increase in the number of functional HER-2/neu-specific CTLs, and the increased tumor infiltration of both CD4+ and CD8+ T cells, depletion of which abolishes the antitumor effect of the HER-2/neu DNA vaccine-AC therapy. Our results further indicate that the treatment of mice with AC enhanced DC activation and production of Th1-activating cytokines (e.g. IL-12, and IFN-alpha) in the draining lymph nodes, which were sufficient to directly stimulate T cell proliferation and higher IFN-gamma production in response to ErbB2. Overall, these results clearly demonstrate that AC represents a promising immunomodulatory adjuvant that could enhance the therapeutic potency of HER-2/neu DNA vaccines in cancer therapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antrodia , Carcinoma/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Receptor, ErbB-2/immunology , Urinary Bladder Neoplasms/immunology , Animals , Apoptosis/drug effects , Carcinoma/drug therapy , Carcinoma/pathology , Cell Extracts/administration & dosage , Cell Extracts/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Female , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Receptor, ErbB-2/genetics , Th1 Cells/immunology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Vaccines, DNA
14.
Kaohsiung J Med Sci ; 24(11): 577-90, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19239991

ABSTRACT

Cutaneous T cell lymphoma (CTCL) is a clonal epidermotropic malignancy of memory T cells primarily involving the skin. However, the mechanisms governing migration of CTCL cells have not been fully clarified. It has been shown that certain chemokine receptors are upregulated in CTCL cells, but it remains unanswered whether these chemokine receptors play a critical role in the migration dynamics of CTCL. Using cell lines originally derived from patients with different subtypes of CTCL, we have shown higher CCR4 expression in the line derived from the mycosis fungoides (MJ), compared with the line derived from Sezary syndrome (Hut78). In specific responses to CCL22 (a CCR4 ligand) treatments, MJ cells showed significant chemotactic migration, enhanced activation and adhesion of certain integrins (CD49d and CD29) in vitro, while the control cells (Hut78, CD4+CD45RO+ memory T cells, and Jurkat cells) did not. Furthermore, compared with Hut78 cells, MJ cells manifested significantly more transendothelial migration in responses to treatments with either CCL22 or conditioned medium from dendritic cells in vitro. These results provide further dynamic evidence, in line with the multistep cascade paradigm for leukocyte transendothelial migration, to support a critical role for CCR4 in CTCL migration.


Subject(s)
Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Receptors, CCR4/metabolism , Skin Diseases/metabolism , Skin Diseases/pathology , Cell Adhesion , Chemokine CCL22/metabolism , Chemokines/metabolism , Chemokines, CXC , Chemotaxis , Dendritic Cells/metabolism , Endosomes/metabolism , Humans , Integrin alpha4beta1/metabolism , Ligands , Mycosis Fungoides/metabolism , Protein Binding , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL