Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.221
Filter
1.
Sci Rep ; 14(1): 23231, 2024 10 05.
Article in English | MEDLINE | ID: mdl-39369019

ABSTRACT

Chrysin is a natural flavonoid with anti-cancer effects. Despite its beneficial effects, little information is available regarding its immunogenic cell death (ICD) properties. In this work, we hypothesized that chrysin can potentiate radiotherapy(RT)-induced immunogenicity in melanoma cell line (B16-F10). We examined the effects of chrysin alone and in combination with radiation on ICD induction in B16-F10 cells. Cell viability was assessed using an MTT assay. Cell apoptosis and calreticulin (CRT) exposure were determined using flow cytometry. Western blotting and ELISA assay were employed to examine changes in protein expression. Combination therapy exhibited a synergistic effect, with an optimum combination index of 0.66. The synergistic anti-cancer effect correlated with increased cell apoptosis in cancer cells. Compared to the untreated control, chrysin alone and in combination with RT induced higher levels of DAMPs, such as CRT, HSP70, HMGB1, and ATP. The protein expression of p-STAT3/STAT3 and PD-L1 was reduced in B16-F10 cells exposed to chrysin alone and in combination with RT. Conditioned media from B16-F10 cells exposed to mono-and combination treatments elicited IL-12 secretion in dendritic cells (DCs), inducing a Th1 response. Our findings revealed that chrysin could induce ICD and intensify the RT-induced immunogenicity.


Subject(s)
Apoptosis , Calreticulin , Flavonoids , Immunogenic Cell Death , Melanoma, Experimental , Flavonoids/pharmacology , Animals , Immunogenic Cell Death/drug effects , Mice , Cell Line, Tumor , Calreticulin/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Apoptosis/drug effects , HMGB1 Protein/metabolism , STAT3 Transcription Factor/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , B7-H1 Antigen/metabolism , Interleukin-12/metabolism , HSP70 Heat-Shock Proteins/metabolism , Adenosine Triphosphate/metabolism
2.
Sci Adv ; 10(40): eado8366, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365866

ABSTRACT

Despite our increasing understanding of macrophage heterogeneity, drivers of macrophage phenotypic and functional polarization in the microenvironment are not fully elucidated. Here, our single-cell RNA sequencing data identify a subpopulation of macrophages expressing high levels of the phagocytic receptor MER proto-oncogene tyrosine kinase (MerTK+ macrophages), which is closely associated with melanoma progression and immunotherapy resistance. Adoptive transfer of the MerTK+ macrophages into recipient mice notably accelerated tumor growth regardless of macrophage depletion. Mechanistic studies further revealed that ALK And LTK Ligand 1 (ALKAL1), a target gene of aryl hydrocarbon receptor (AhR), facilitated MerTK phosphorylation, resulting in heightened phagocytic activity of MerTK+ macrophages and their subsequent polarization toward an immunosuppressive phenotype. Specifically targeted delivery of AhR antagonist to tumor-associated macrophages with mannosylated micelles could suppress MerTK expression and improved the therapeutic efficacy of anti-programmed cell death ligand 1 therapy. Our findings shed light on the regulatory mechanism of MerTK+ macrophages and provide strategies for improving the efficacy of melanoma immunotherapy.


Subject(s)
Immunotherapy , Macrophages , Melanoma , Receptors, Aryl Hydrocarbon , c-Mer Tyrosine Kinase , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , Animals , Mice , Melanoma/therapy , Melanoma/immunology , Melanoma/pathology , Melanoma/metabolism , Immunotherapy/methods , Receptors, Aryl Hydrocarbon/metabolism , Macrophages/metabolism , Macrophages/immunology , Tumor Microenvironment/immunology , Humans , Disease Progression , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Proto-Oncogene Mas , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Phosphorylation , Drug Resistance, Neoplasm
3.
Cell Commun Signal ; 22(1): 437, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261954

ABSTRACT

Squalene (SQ) is a well-known antioxidant and anti-inflammatory agent that provides promising anti-aging and UV-protective roles on human skin. However, its strong hydrophobic nature, accompanied by issues such as poor solubility and limited tissue permeation, has created challenges for scientists to investigate its untapped potential in more complex conditions, including cancer progression. The present study assessed the potent anti-metastatic properties of a newly synthesized amphiphilic ethylene glycol SQ derivative (SQ-diEG) in melanoma, the most fatal skin cancer. In vitro and in vivo experiments have discovered that SQ-diEG may exert its potential on melanoma malignancy through the mitochondria-mediated caspase activation apoptotic signaling pathway. The potent anti-metastatic effect of SQ-diEG was observed in vitro using highly proliferative and aggressive melanoma cells. Administration of SQ-diEG (25 mg/kg) significantly decreased the tumor burden on the lung and inhibited the metastasis-associated proteins and gene markers in B16F10 lung colonization mice model. Furthermore, global gene profiling also revealed a promising role of SQ-diEG in tumor microenvironment. We anticipated that the amphiphilic nature of the SQ compound bearing ethylene glycol oligomers could potentially augment its ability to reach the pathology site, thus enhancing its therapeutic potential in melanoma.


Subject(s)
Melanoma , Squalene , Animals , Mice , Squalene/chemistry , Squalene/pharmacology , Humans , Cell Line, Tumor , Melanoma/pathology , Melanoma/drug therapy , Mice, Inbred C57BL , Apoptosis/drug effects , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Neoplasm Metastasis , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Ethers/pharmacology , Ethers/chemistry , Cell Proliferation/drug effects , Tumor Microenvironment/drug effects , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry
4.
J Med Chem ; 67(18): 16195-16208, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39264254

ABSTRACT

In this paper, three new iridium(III) complexes: [Ir(piq)2(DFIPP)]PF6 (piq = deprotonated 1-phenylisoquinoline, DFIPP = 3,4-difluoro-2-(1H-imidazo[4,5-f][1,10]phenenthrolin-2-yl)phenol, 3a), [Ir(bzq)2(DFIPP)]PF6 (bzq = deprotonated benzo[h]quinoline, 3b), and [Ir(ppy)2(DFIPP)]PF6 (ppy = deprotonated 1-phenylpyridine, 3c), were synthesized and characterized. The complexes were found to be nontoxic to tumor cells via 3-(4,5-dimethylthiazole-2-yl)-diphenyltetrazolium bromide (MTT) assay. Surprisingly, its liposome-entrapped complexes 3alip, 3blip, and 3clip on B16 cells showed strong cytotoxicity (IC50 = 13.6 ± 2.8, 9.6 ± 1.1, and 18.9 ± 2.1 µM). Entry of 3alip, 3blip, and 3clip into B16 cells decreases mitochondrial membrane potential, regulates Bcl-2 family proteins, releases cytochrome c, triggers caspase family cascade reaction, and induces apoptosis. In addition, we also found that 3alip, 3blip, and 3clip triggered ferroptosis and autophagy. In vivo studies demonstrated that 3blip inhibited melanoma growth in C57 mice with a high inhibitory rate of 83.95%, and no organic damage was found in C57 mice.


Subject(s)
Antineoplastic Agents , Apoptosis , Coordination Complexes , Iridium , Liposomes , Iridium/chemistry , Iridium/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mice , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Humans , Mice, Inbred C57BL , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Membrane Potential, Mitochondrial/drug effects
5.
Carbohydr Polym ; 344: 122466, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218533

ABSTRACT

Traditional fungi ß-glucan commonly possesses high molecular weight with poor water solubility, which remains significant challenge in the drug development and medical application. Water-soluble ß-glucan with high molecular weight (dHSCG) of 560 kDa, low molecular weight (dLSCG) of 60 kDa, and sulfated derivative (SCGS) with a molecular weight of 146 kDa and sulfate degree at 2.04 were obtained through well-controlled degradation and sulfated modification from Saccharomyces cerevisiae in this study. The structural characteristics were confirmed as ß-1,3/6-glucan by FT-IR and NMR spectroscopy. Carbohydrate microarrays and surface plasmon resonance revealed distinct and contrasting binding affinities between the natural ß-glucans and sulfated derivatives. SCGS exhibited strong binding to FGF and VEGF, while natural ß-glucan showed no response, suggesting its potential as a novel antitumor agent. Moreover, SCGS significantly inhibited the migration rate of the highly metastatic melanoma (B16F10) cells. The lung metastasis mouse model also demonstrated that SCGS significantly reduced and eliminated the nodules, achieving an inhibition rate of 86.7% in vivo, with a dramatic improvement in IFN-α, TNF-α, and IL-1ß levels. Through analysis of protein content and distribution in lung tissues, the anti-tumor and anti-metastasis mechanism of SCGS involves the regulation of degrading enzymes to protect extracellular matrix (ECM), as well as the reduction of angiogenic factor release. These findings provide a foundation for exploring the potential of SCGS in the development of new anti-tumor and anti-metastasis drugs and open up a new field in cancer research.


Subject(s)
Antineoplastic Agents , Saccharomyces cerevisiae , Solubility , beta-Glucans , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , beta-Glucans/chemistry , beta-Glucans/pharmacology , Water/chemistry , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Sulfates/chemistry , Cell Movement/drug effects , Humans
6.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225102

ABSTRACT

Tumor reliance on glycolysis is a hallmark of cancer. Immunotherapy is more effective in controlling glycolysis-low tumors lacking lactate dehydrogenase (LDH) due to reduced tumor lactate efflux and enhanced glucose availability within the tumor microenvironment (TME). LDH inhibitors (LDHi) reduce glucose uptake and tumor growth in preclinical models, but their impact on tumor-infiltrating T cells is not fully elucidated. Tumor cells have higher basal LDH expression and glycolysis levels compared with infiltrating T cells, creating a therapeutic opportunity for tumor-specific targeting of glycolysis. We demonstrate that LDHi treatment (a) decreases tumor cell glucose uptake, expression of the glucose transporter GLUT1, and tumor cell proliferation while (b) increasing glucose uptake, GLUT1 expression, and proliferation of tumor-infiltrating T cells. Accordingly, increasing glucose availability in the microenvironment via LDH inhibition leads to improved tumor-killing T cell function and impaired Treg immunosuppressive activity in vitro. Moreover, combining LDH inhibition with immune checkpoint blockade therapy effectively controls murine melanoma and colon cancer progression by promoting effector T cell infiltration and activation while destabilizing Tregs. Our results establish LDH inhibition as an effective strategy for rebalancing glucose availability for T cells within the TME, which can enhance T cell function and antitumor immunity.


Subject(s)
Glucose , L-Lactate Dehydrogenase , Tumor Microenvironment , Animals , Mice , Glucose/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/immunology , Humans , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/immunology , Glucose Transporter Type 1/genetics , Cell Line, Tumor , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Glycolysis/drug effects , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Immunotherapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use
7.
J Clin Invest ; 134(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39286984

ABSTRACT

T cell-based immunotherapies are a promising therapeutic approach for multiple malignancies, but their efficacy is limited by tumor hypoxia arising from dysfunctional blood vessels. Here, we report that cell-intrinsic properties of a single vascular component, namely the pericyte, contribute to the control of tumor oxygenation, macrophage polarization, vessel inflammation, and T cell infiltration. Switching pericyte phenotype from a synthetic to a differentiated state reverses immune suppression and sensitizes tumors to adoptive T cell therapy, leading to regression of melanoma in mice. In melanoma patients, improved survival is correlated with enhanced pericyte maturity. Importantly, pericyte plasticity is regulated by signaling pathways converging on Rho kinase activity, with pericyte maturity being inducible by selective low-dose therapeutics that suppress pericyte MEK, AKT, or notch signaling. We also show that low-dose targeted anticancer therapy can durably change the tumor microenvironment without inducing adaptive resistance, creating a highly translatable pathway for redosing anticancer targeted therapies in combination with immunotherapy to improve outcome.


Subject(s)
Pericytes , Animals , Pericytes/immunology , Pericytes/metabolism , Pericytes/pathology , Mice , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Immunotherapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Phenotype , Melanoma/immunology , Melanoma/therapy , Melanoma/pathology , Melanoma/drug therapy , Cell Line, Tumor , Immune Tolerance/drug effects
8.
Cancer Immunol Immunother ; 73(11): 218, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235510

ABSTRACT

Epigenetic modifications to DNA and chromatin control oncogenic and tumor-suppressive mechanisms in melanoma. Ezh2, the catalytic component of the Polycomb Repressive Complex 2 (PRC2), which mediates methylation of lysine 27 on histone 3 (H3K27me3), can regulate both melanoma initiation and progression. We previously found that mutant Ezh2Y641F interacts with the immune regulator Stat3 and together they affect anti-tumor immunity. However, given the numerous downstream targets and pathways affected by Ezh2, many mechanisms that determine its oncogenic activity remain largely unexplored. Using genetically engineered mouse models, we further investigated the role of pathways downstream of Ezh2 in melanoma carcinogenesis and identified significant enrichment in several autophagy signatures, along with increased expression of autophagy regulators, such as Atg7. In this study, we investigated the effect of Atg7 on melanoma growth and tumor immunity within the context of a wild-type or Ezh2Y641F epigenetic state. We found that the Atg7 locus is controlled by multiple Ezh2 and Stat3 binding sites, Atg7 expression is dependent on Stat3 expression, and that deletion of Atg7 slows down melanoma cell growth in vivo, but not in vitro. Atg7 deletion also results in increased CD8 + T cells in Ezh2Y641F melanomas and reduced myelosuppressive cell infiltration in the tumor microenvironment, particularly in Ezh2WT melanomas, suggesting a strong immune system contribution in the role of Atg7 in melanoma progression. These findings highlight the complex interplay between genetic mutations, epigenetic regulators, and autophagy in shaping tumor immunity in melanoma.


Subject(s)
Autophagy-Related Protein 7 , Enhancer of Zeste Homolog 2 Protein , STAT3 Transcription Factor , Animals , STAT3 Transcription Factor/metabolism , Mice , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Gene Expression Regulation, Neoplastic , Melanoma/immunology , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Epigenesis, Genetic , Cell Line, Tumor , Humans , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism
9.
ACS Appl Mater Interfaces ; 16(37): 49164-49175, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39252418

ABSTRACT

Focused on the newly secreted tumorous exosomes during melanoma immunotherapy, this work has pioneered an ultra-sensitive spatiotemporal-specific exosome detection strategy, leveraging advanced exosomal membrane engineering techniques. The proposed strategy harnesses the power of amplified lanthanide luminescence signals on these exosomes, enabling precise and real-time monitoring of the efficacy of melanoma immunotherapy. The methodology comprises two pivotal steps. Initially, Ac4ManNAz-associated metabolic labeling is employed to evolve azide groups onto the membranes of newly secreted exosomes with remarkable selectivity. These azide groups serve as versatile clickable artificial tags, enabling the precise identification of melanoma exosomes emerging during immunotherapy. Subsequently, lanthanide-nanoparticle-functionalized polymer chains are controllably grafted onto the exosome surfaces through click chemistry and in situ Fenton-RAFT polymerization, serving as robust signal amplifiers. When integrated with time-resolved fluorescence detection, this strategy yields detection signals with an exceptionally high signal-to-noise ratio, enabling ultra-sensitive detection of PD-L1 antigen expression levels on the spatiotemporal-specific exosomes. The detection strategy boasts a wide linear concentration range spanning from 1.7 × 104 to 1.7 × 109 particles/mL, with a remarkable theoretical detection limit of 1.28 × 103 particles/mL. The remarkable enhancements in detection sensitivity and accuracy facilitate the evaluation of the efficacy of immunotherapeutic interventions in the mouse B16 melanoma model, notably revealing a substantial disparity in PD-L1 levels between immunotherapy-treated and untreated groups (P < 0.01) and further emphasizing the cumulative therapeutic effect that intensifies with repeated treatments (P < 0.001).


Subject(s)
Exosomes , Immunotherapy , Lanthanoid Series Elements , Exosomes/chemistry , Exosomes/metabolism , Animals , Mice , Lanthanoid Series Elements/chemistry , Melanoma/therapy , Melanoma/metabolism , Melanoma/immunology , Melanoma/pathology , Luminescence , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Cell Line, Tumor , Humans , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Nanoparticles/chemistry
10.
Nat Commun ; 15(1): 8354, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333093

ABSTRACT

T cell inhibitory mechanisms prevent autoimmune reactions, while cancer immunotherapy aims to remove these inhibitory signals. Chronic ultraviolet (UV) exposure attenuates autoimmunity through promotion of poorly understood immune-suppressive mechanisms. Here we show that mice with subcutaneous melanoma are not responsive to anti-PD1 immunotherapy following chronic UV irradiation, given prior to tumor injection, due to the suppression of T cell killing ability in skin-draining lymph nodes. Using mass cytometry and single-cell RNA-sequencing analyzes, we discover that skin-specific, UV-induced suppression of T-cells killing activity is mediated by upregulation of a Ly6ahigh T-cell subpopulation. Independently of the UV effect, Ly6ahigh T cells are induced by chronic type-1 interferon in the tumor microenvironment. Treatment with an anti-Ly6a antibody enhances the anti-tumoral cytotoxic activity of T cells and reprograms their mitochondrial metabolism via the Erk/cMyc axis. Treatment with an anti-Ly6a antibody inhibits tumor growth in mice resistant to anti-PD1 therapy. Applying our findings in humans could lead to an immunotherapy treatment for patients with resistance to existing treatments.


Subject(s)
Antigens, Ly , CD8-Positive T-Lymphocytes , Immunotherapy , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Antigens, Ly/metabolism , Antigens, Ly/immunology , Mice , Immunotherapy/methods , Tumor Microenvironment/immunology , Mice, Inbred C57BL , Cell Line, Tumor , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Female , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Skin Neoplasms/pathology , Mitochondria/metabolism , Melanoma/immunology , Melanoma/therapy , Interferon Type I/metabolism
11.
Cells ; 13(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39329721

ABSTRACT

Impaired tumor cell antigen presentation contributes significantly to immune evasion. This study identifies Berbamine hydrochloride (Ber), a compound derived from traditional Chinese medicine, as an effective inhibitor of autophagy that enhances antigen presentation in tumor cells. Ber increases MHC-I-mediated antigen presentation in melanoma cells, improving recognition and elimination by CD8+ T cells. Mutation of Atg4b, which blocks autophagy, also raises MHC-I levels on the cell surface, and further treatment with Ber under these conditions does not increase MHC-I, indicating Ber's role in blocking autophagy to enhance MHC-I expression. Additionally, Ber treatment leads to the accumulation of autophagosomes, with elevated levels of LC3-II and p62, suggesting a disrupted autophagic flux. Fluorescence staining and co-localization analyses reveal that Ber likely inhibits lysosomal acidification without hindering autophagosome-lysosome fusion. Importantly, Ber treatment suppresses melanoma growth in mice and enhances CD8+ T cell infiltration, supporting its therapeutic potential. Our findings demonstrate that Ber disturbs late-stage autophagic flux through abnormal lysosomal acidification, enhancing MHC-I-mediated antigen presentation and curtailing tumor immune escape.


Subject(s)
Autophagy , Benzylisoquinolines , Melanoma , Tumor Escape , Autophagy/drug effects , Animals , Mice , Cell Line, Tumor , Humans , Tumor Escape/drug effects , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Melanoma/drug therapy , Melanoma/pathology , Melanoma/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Antigen Presentation/drug effects , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Mice, Inbred C57BL , Autophagosomes/metabolism , Autophagosomes/drug effects , Lysosomes/metabolism , Lysosomes/drug effects , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Cysteine Endopeptidases
12.
Nano Lett ; 24(34): 10664-10673, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39140448

ABSTRACT

Here we report a brand-new bioactive polymer featuring sulfonium moieties that exhibits the capability of inducing immunogenic cell death (ICD) for anticancer therapy. The optimized polysulfonium presents a wide spectrum of potent anticancer activity and remarkable selectivity. In-depth mechanistic studies reveal that the polymer exerts its cytotoxic effects on cancer cells through a membrane-disrupting mechanism. This further initiates the release of a plethora of damage-associated molecular patterns, effectively triggering ICD and resulting in systemic anticancer immune responses. Notably, the compound demonstrated significant efficacy in suppressing tumor growth in the B16-F10 melanoma tumor model. Furthermore, it exhibits robust immune memory effects, effectively suppressing tumor recurrence and metastasis in both the rechallenge model and the lung metastatic tumor model. To the best of our knowledge, the study represents the pioneering exportation of cationic polysulfoniums, showcasing not only their remarkable safety and efficacy against primary tumors but also their unique ability in activating long-term immune memory.


Subject(s)
Antineoplastic Agents , Immunogenic Cell Death , Polymers , Animals , Immunogenic Cell Death/drug effects , Mice , Humans , Cell Line, Tumor , Polymers/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Sulfonium Compounds/chemistry , Sulfonium Compounds/pharmacology , Sulfonium Compounds/therapeutic use , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology
13.
ACS Nano ; 18(34): 23797-23811, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39140567

ABSTRACT

Cancer immunotherapy has emerged as a promising approach to cancer treatment in recent years. The physical and chemical properties of nanocarriers are critical factors that regulate the immune activation of antigen-presenting cells (APCs) in the tumor microenvironment (TME). Herein, we extensively investigated the behavior of liposome nanoparticles (Lipo-NPs) with different elasticities, focusing on their interaction with immune cells and their transport mechanisms from tumors to tumor-draining lymph nodes (tdLNs). Successfully preparing Lipo-NPs with distinct elastic properties, their varied behaviors were observed, concerning immune cell interaction. Soft Lipo-NPs exhibited an affinity to cell membranes, while those with medium elasticity facilitated the cargo delivery to macrophages through membrane fusion. Conversely, hard Lipo-NPs enter macrophages via classical cellular uptake pathways. Additionally, it was noted that softer Lipo-NPs displayed superior transport to tdLNs in vivo, attributed to their deformable nature with lower elasticity. As a result, the medium elastic Lipo-NPs with agonists (cGAMP), by activating the STING pathway and enhancing transport to tdLNs, promoted abundant infiltration of tumor-infiltrating lymphocytes (TILs), leading to notable antitumor effects and extended survival in a melanoma mouse model. Furthermore, this study highlighted the potential synergistic effect of medium elasticity Lipo-NPs with immune checkpoint blockade (ICB) therapy in preventing tumor immune evasion. These findings hold promise for guiding immune-targeted delivery systems in cancer immunotherapy, particularly in vaccine design for tdLNs targeting and eradicating metastasis within tdLNs.


Subject(s)
Elasticity , Immunotherapy , Liposomes , Liposomes/chemistry , Animals , Mice , Mice, Inbred C57BL , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Humans , Female , Melanoma, Experimental/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Cell Line, Tumor
14.
ACS Nano ; 18(34): 23289-23300, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39151414

ABSTRACT

mRNA vaccines have been revolutionizing disease prevention and treatment. However, their further application is hindered by inflammatory side effects, primarily caused by delivery systems such as lipid nanoparticles (LNPs). In response to this issue, we prepared cationic lipids (mLPs) derived from mildronate, a small-molecule drug, and subsequently developed the LNP (mLNP-69) comprising a low dose of mLP. Compared with the LNP (sLNP) based on SM-102, a commercially available ionizable lipid, mLNP-69 ensures effective mRNA delivery while significantly reducing local inflammation. In preclinical prophylactic and therapeutic B16-OVA melanoma models, mLNP-69 demonstrated successful mRNA cancer vaccine delivery in vivo, effectively preventing tumor occurrence or impeding tumor progression. The results suggest that the cationic lipids derived from mildronate, which exhibit efficient delivery capabilities and minimal inflammatory side effects, hold great promise for clinical application.


Subject(s)
Inflammation , Lipids , Animals , Mice , Lipids/chemistry , Inflammation/prevention & control , Nanoparticles/chemistry , Mice, Inbred C57BL , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , mRNA Vaccines , RNA, Messenger/genetics , Female , Melanoma, Experimental/pathology
15.
J Exp Clin Cancer Res ; 43(1): 214, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39090759

ABSTRACT

BACKGROUND: Melanoma progression is based on a close interaction between cancer cells and immune cells in the tumor microenvironment (TME). Thus, a better understanding of the mechanisms controlling TME dynamics and composition will help improve the management of this dismal disease. Work from our and other groups has reported the requirement of an active Hedgehog-GLI (HH-GLI) signaling for melanoma growth and stemness. However, the role of the downstream GLI1 transcription factor in melanoma TME remains largely unexplored. METHODS: The immune-modulatory activity of GLI1 was evaluated in a syngeneic B16F10 melanoma mouse model assessing immune populations by flow cytometry. Murine polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were differentiated from bone marrow cells and their immunosuppressive ability was assessed by inhibition of T cells. Conditioned media (CM) from GLI1-overexpressing mouse melanoma cells was used to culture PMN-MDSCs, and the effects of CM were evaluated by Transwell invasion assay and T cell inhibition. Cytokine array analysis, qPCR and chromatin immunoprecipitation were performed to explore the regulation of CX3CL1 expression by GLI1. Human monocyte-derived dendritic cells (moDCs) were cultured in CM from GLI1-silenced patient-derived melanoma cells to assess their activation and recruitment. Blocking antibodies anti-CX3CL1, anti-CCL7 and anti-CXCL8 were used for in vitro functional assays. RESULTS: Melanoma cell-intrinsic activation of GLI1 promotes changes in the infiltration of immune cells, leading to accumulation of immunosuppressive PMN-MDSCs and regulatory T cells, and to decreased infiltration of dendric cells (DCs), CD8 + and CD4 + T cells in the TME. In addition, we show that ectopic expression of GLI1 in melanoma cells enables PMN-MDSC expansion and recruitment, and increases their ability to inhibit T cells. The chemokine CX3CL1, a direct transcriptional target of GLI1, contributes to PMN-MDSC expansion and recruitment. Finally, silencing of GLI1 in patient-derived melanoma cells promotes the activation of human monocyte-derived dendritic cells (moDCs), increasing cytoskeleton remodeling and invasion ability. This phenotype is partially prevented by blocking the chemokine CCL7, but not CXCL8. CONCLUSION: Our findings highlight the relevance of tumor-derived GLI1 in promoting an immune-suppressive TME, which allows melanoma cells to evade the immune system, and pave the way for the design of new combination treatments targeting GLI1.


Subject(s)
Melanoma , Myeloid-Derived Suppressor Cells , Tumor Microenvironment , Zinc Finger Protein GLI1 , Animals , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Mice , Humans , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Melanoma/pathology , Melanoma/metabolism , Melanoma/immunology , Melanoma/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Inbred C57BL
16.
ACS Chem Biol ; 19(8): 1820-1835, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39099090

ABSTRACT

Neuropilin-1 acts as a coreceptor with vascular endothelial growth factor receptors to facilitate binding of its ligand, vascular endothelial growth factor. Neuropilin-1 also binds to heparan sulfate, but the functional significance of this interaction has not been established. A combinatorial library screening using heparin oligosaccharides followed by molecular dynamics simulations of a heparin tetradecasaccharide suggested a highly conserved binding site composed of amino acid residues extending across the b1 and b2 domains of murine neuropilin-1. Mutagenesis studies established the importance of arginine513 and lysine514 for binding of heparin to a recombinant form of Nrp1 composed of the a1, a2, b1, and b2 domains. Recombinant Nrp1 protein bearing R513A,K514A mutations showed a significant loss of heparin-binding, heparin-induced dimerization, and heparin-dependent thermal stabilization. Isothermal calorimetry experiments suggested a 1:2 complex of heparin tetradecasaccharide:Nrp1. To study the impact of altered heparin binding in vivo, a mutant allele of Nrp1 bearing the R513A,K514A mutations was created in mice (Nrp1D) and crossbred to Nrp1+/- mice to examine the impact of altered heparan sulfate binding. Analysis of tumor formation showed variable effects on tumor growth in Nrp1D/D mice, resulting in a frank reduction in tumor growth in Nrp1D/- mice. Expression of mutant Nrp1D protein was normal in tissues, suggesting that the reduction in tumor growth was due to the altered binding of heparin/heparan sulfate to neuropilin-1. These findings suggest that the interaction of neuropilin-1 with heparan sulfate modulates its stability and its role in tumor formation and growth.


Subject(s)
Heparitin Sulfate , Neuropilin-1 , Neuropilin-1/metabolism , Neuropilin-1/genetics , Neuropilin-1/chemistry , Animals , Heparitin Sulfate/metabolism , Mice , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Protein Binding , Binding Sites , Mice, Inbred C57BL , Heparin/metabolism , Heparin/chemistry , Molecular Dynamics Simulation , Mutation
17.
Nat Commun ; 15(1): 6613, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098861

ABSTRACT

Tumour-host immune interactions lead to complex changes in the tumour microenvironment (TME), impacting progression, metastasis and response to therapy. While it is clear that cancer cells can have the capacity to alter immune landscapes, our understanding of this process is incomplete. Herein we show that endocytic trafficking at the plasma membrane, mediated by the small GTPase ARF6, enables melanoma cells to impose an immunosuppressive TME that accelerates tumour development. This ARF6-dependent TME is vulnerable to immune checkpoint blockade therapy (ICB) but in murine melanoma, loss of Arf6 causes resistance to ICB. Likewise, downregulation of ARF6 in patient tumours correlates with inferior overall survival after ICB. Mechanistically, these phenotypes are at least partially explained by ARF6-dependent recycling, which controls plasma membrane density of the interferon-gamma receptor. Collectively, our findings reveal the importance of endomembrane trafficking in outfitting tumour cells with the ability to shape their immune microenvironment and respond to immunotherapy.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Cell Membrane , Immune Checkpoint Inhibitors , Melanoma , Tumor Microenvironment , Tumor Microenvironment/immunology , Animals , Humans , Mice , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Melanoma/immunology , Cell Line, Tumor , Cell Membrane/metabolism , Interferon gamma Receptor , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Protein Transport , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/genetics , Mice, Inbred C57BL , Female
18.
Skin Res Technol ; 30(8): e13869, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39171844

ABSTRACT

BACKGROUND: As a medicinal and food homologous plant, Rosa damascena is not only highly ornamental, but also rich in a variety of active ingredients such as polyphenols and flavonoids. It is widely used in cosmetics, food and pharmaceutical industries. OBJECTIVE: To study the in vitro efficacy of Rosa damascena solid state fermentation liquid (RDF) and water extract (RDE). METHODS: Firstly, the effect of RDF and RDE on the proliferation rate of B16F10 cells was detected by CCK-8 method, and the melanin content was measured by sodium hydroxide lysis method to evaluate the whitening effect of them. Finally, the antioxidant, anti-wrinkling and soothing effects of RDF and RDE were evaluated by biochemical methods in vitro. RESULTS: RDF and RDE within a certain concentration range (0.05%-0.5%) had no effect on the proliferation of B16F10 cells. Compared with Rosa damascena extract (RDE), RDF showed significant effects on bleaching, antioxidant, anti-wrinkling and soothing, among which 0.5% RDF showed the best effect. CONCLUSION: Both RDF and RDE at a certain concentration have effect on skin care in vitro, but the effect of RDF is more significant than that of RDE.


Subject(s)
Antioxidants , Cell Proliferation , Fermentation , Plant Extracts , Rosa , Rosa/chemistry , Plant Extracts/pharmacology , Mice , Animals , Cell Proliferation/drug effects , Antioxidants/pharmacology , Skin Care/methods , Water/chemistry , Skin Aging/drug effects , Melanins , Cell Line, Tumor , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology
19.
ACS Appl Bio Mater ; 7(8): 5057-5075, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39115261

ABSTRACT

Melanoma has gained considerable attention due to its high mortality and morbidity rate worldwide. The currently available treatment options are associated with several limitations such as nonspecificity, drug resistance, easy clearance, low efficacy, toxicity-related issues, etc. To this end, nanotechnology has garnered significant attention for the treatment of melanoma. In the present manuscript, we have demonstrated the in vitro and in vivo anticancer activity of silver nitroprusside nanoparticles (abbreviated as AgNNPs) against melanoma. The AgNNPs exhibit cytotoxicity against B16F10 cells, which has been investigated by several in vitro experiments including [methyl 3H]-thymidine incorporation assay, cell cycle and apoptosis analysis by flow cytometry, and ROS generation through DCFDA, DHE, and DAF2A reagents. Further, the internalization of nanoparticles was determined by ICPOES analysis, while their colocalization was analyzed by confocal microscopy. Additionally, JC-1 staining is performed to examine mitochondrial membrane potential (MMP). Cytoskeleton integrity was observed by phalloidin staining. Expression of different markers (Ki-67, cytochrome c, and E-cadherin) was checked using an immunofluorescence assay. The in vivo therapeutic efficacy of AgNNPs has been validated in the melanoma model established by inoculating B16F10 cells into the dorsal right abdomen of C57BL/6J mice. The intraperitoneal administration of AgNNPs reduced melanoma growth and increased the survivability of tumor-bearing mice. The in vivo immunofluorescence studies (Ki-67, CD31, and E-cadherin) and TUNEL assay support the inhibitory and apoptotic nature of AgNNPs toward melanoma, respectively. Furthermore, the various signaling pathways and molecular mechanisms involved in anticancer activity are evaluated by Western blot analysis. These findings altogether demonstrate the promising anticancer potential of AgNNPs toward melanoma.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Drug Screening Assays, Antitumor , Mice, Inbred C57BL , Nitroprusside , Silver , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Nitroprusside/pharmacology , Nitroprusside/chemistry , Apoptosis/drug effects , Silver/chemistry , Silver/pharmacology , Cell Proliferation/drug effects , Particle Size , Materials Testing , Melanoma/drug therapy , Melanoma/pathology , Metal Nanoparticles/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Nanoparticles/chemistry , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism
20.
J Dermatol Sci ; 115(3): 121-129, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127591

ABSTRACT

BACKGROUND: Dysregulation of melanogenesis contributes to the development of skin hyperpigmentation diseases, which poses a treatment challenge. Following the establishment of CRTC3 screening methods to explore small molecules inhibiting melanogenesis for the topical treatment of hyperpigmentation diseases, we identified a candidate molecule, semaxanib. OBJECTIVE: To explore the antimelanogenic effects of semaxanib, a vascular endothelial growth factor receptor (VEGFR) 2 inhibitor, for potential applications in hyperpigmentation management and to unravel the role of VEGF signaling in melanocyte biology by investigating mechanism of action of semaxanib. METHODS: Mouse-derived spontaneously immortalized melanocytes, B16F10, and normal human primary epidermal melanocytes cells were treated with semaxanib, and cellular responses were assessed using cell viability assays and melanin content measurements. Molecular mechanisms were investigated using transcriptional activity assays, reverse-transcription polymerase chain reaction, and immunoblotting analysis. In vivo studies were conducted using an epidermis-humanized transgenic mouse model and ex vivo human skin tissues. RESULTS: Semaxanib ameliorated melanin content in cultured melanocytes by downregulating the expression of melanogenesis-associated genes by suppressing the CRTC3/microphthalmia-associated transcription factors. Topical application of semaxanib reduced melanin accumulation in the ultraviolet B-stimulated ex vivo human epidermis and tail of K14-stem cell factor transgenic mice. Mechanistically, the antimelanogenic effect induced by semaxanib was associated with SIK2-CRTC3-MITF rather than VEGF signaling in melanocytes. CONCLUSION: Semaxanib emerges as a promising candidate for the development of therapeutics for hyperpigmentation, potentially working independently of VEGF signaling in human melanocytes.


Subject(s)
Melanins , Melanocytes , Microphthalmia-Associated Transcription Factor , Signal Transduction , Transcription Factors , Vascular Endothelial Growth Factor A , Animals , Melanocytes/drug effects , Melanocytes/metabolism , Humans , Melanins/biosynthesis , Melanins/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Signal Transduction/drug effects , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Indoles/pharmacology , Hyperpigmentation/drug therapy , Mice, Transgenic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Cells, Cultured , Melanogenesis
SELECTION OF CITATIONS
SEARCH DETAIL