Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
J Infect Public Health ; 16(1): 42-54, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36470006

ABSTRACT

BACKGROUND: The novel coronavirus disease-2019 (COVID-19) that emerged in China, is an extremely contagious and pathogenic viral infection caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) that has sparked a global pandemic. The few and limited availability of approved therapeutic agents or vaccines is of great concern. Urgently, Remdesivir, Nirmatrelvir, Molnupiravir, and some phytochemicals including polyphenol, flavonoid, alkaloid, and triterpenoid are applied to develop as repurposing drugs against the SARS-CoV-2 invasion. METHODS: This study was conducted to perform molecular docking and absorption, distribution, metabolism, excretion and toxicity (ADMET) analysis of the potential phytocompounds and repurposing drugs against three targets of SARS-CoV-2 proteins (RNA dependent RNA polymerase, RdRp, Endoribonclease, S-protein of ACE2-RBD). RESULTS: The docking data illustrated Arachidonic acid, Rutin, Quercetin, and Curcumin were highly bound with coronavirus polyprotein replicase and Ebolavirus envelope protein. Furthermore, anti- Ebolavirus molecule Remedesivir, anti-HIV molecule Chloroquine, and Darunavir were repurposed with coronavirus polyprotein replicase as well as Ebolavirus envelope protein. The strongest binding interaction of each targets are Rutin with RdRp, Endoribonclease with Amentoflavone, and ACE2-RBD with Epigallocatechin gallate. CONCLUSIONS: Taken altogether, these results shed a light on that phytocompounds have a therapeutic potential for the treatment of anti-SARS-CoV-2 may base on multi-target effects or cocktail formulation for blocking viral infection through invasion/activation, transcription/reproduction, and posttranslational cleavage to battle COVID-19 pandemic.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Phytochemicals , Humans , Angiotensin-Converting Enzyme 2 , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/chemistry , Immune Evasion/drug effects , Molecular Docking Simulation , Pandemics , RNA-Dependent RNA Polymerase , Rutin/pharmacology , SARS-CoV-2 , Virus Internalization/drug effects , Virus Replication/drug effects , Phytochemicals/chemistry , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Arachidonic Acid/chemistry , Arachidonic Acid/pharmacology , Quercetin/chemistry , Quercetin/pharmacology , Curcumin/chemistry , Curcumin/pharmacology
2.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35149555

ABSTRACT

SARS-CoV-2 is a highly pathogenic virus that evades antiviral immunity by interfering with host protein synthesis, mRNA stability, and protein trafficking. The SARS-CoV-2 nonstructural protein 1 (Nsp1) uses its C-terminal domain to block the messenger RNA (mRNA) entry channel of the 40S ribosome to inhibit host protein synthesis. However, how SARS-CoV-2 circumvents Nsp1-mediated suppression for viral protein synthesis and if the mechanism can be targeted therapeutically remain unclear. Here, we show that N- and C-terminal domains of Nsp1 coordinate to drive a tuned ratio of viral to host translation, likely to maintain a certain level of host fitness while maximizing replication. We reveal that the stem-loop 1 (SL1) region of the SARS-CoV-2 5' untranslated region (5' UTR) is necessary and sufficient to evade Nsp1-mediated translational suppression. Targeting SL1 with locked nucleic acid antisense oligonucleotides inhibits viral translation and makes SARS-CoV-2 5' UTR vulnerable to Nsp1 suppression, hindering viral replication in vitro at a nanomolar concentration, as well as providing protection against SARS-CoV-2-induced lethality in transgenic mice expressing human ACE2. Thus, SL1 allows Nsp1 to switch infected cells from host to SARS-CoV-2 translation, presenting a therapeutic target against COVID-19 that is conserved among immune-evasive variants. This unique strategy of unleashing a virus' own virulence mechanism against itself could force a critical trade-off between drug resistance and pathogenicity.


Subject(s)
5' Untranslated Regions/genetics , Immune Evasion/genetics , Protein Biosynthesis , SARS-CoV-2/genetics , Viral Nonstructural Proteins/genetics , Animals , Base Sequence , Chlorocebus aethiops , HEK293 Cells , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/drug effects , Mice, Transgenic , Models, Biological , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis/drug effects , SARS-CoV-2/drug effects , Vero Cells , Virus Replication/drug effects
3.
Cell Death Dis ; 12(11): 1033, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34718325

ABSTRACT

Lipopolysaccharide (LPS) as an important inflammatory mediator activates the innate/adaptive immune system. The existence of LPS in pancreatic ductal adenocarcinoma (PDAC) has been reported, however, its biological function in PDAC remains unclear. Here, we demonstrated that circulating and tumoral LPS was significantly increased by intestinal leakage in the orthotopic murine PDAC model, and LPS administration promoted T cell infiltration but exhaustion paradoxically in the subcutaneous murine PDAC model. By bioinformatic analysis, Toll-like receptor 4 (TLR4), LPS receptor, was further found to enrich in immune tolerance signaling in PDAC tissues. Then, a significant positive correlation was found between TLR4 and programmed death ligand-1 (PD-L1) in clinical PDAC tissues, as well as serum LPS and tumoral PD-L1. Meanwhile, LPS stimulation in vitro and in vivo obviously upregulated tumor PD-L1 expression, and effectively promoted cancer cells resistance to T cell cytotoxicity. Mechanistically, the activation of TLR4/MyD88/AKT/NF-κB cascade was found to participate in LPS mediated PD-L1 transcription via binding to its promoter regions, which was enhanced by crosstalk between NF-κB and AKT pathways. Finally, PD-L1 blockade could significantly reverse LPS-induced immune escape, and synergized with LPS treatment. Taken together, LPS can remodel tumor microenvironment, and synergize with PD-L1 blockade to suppress tumor growth, which may be a promising comprehensive strategy for PDAC.


Subject(s)
B7-H1 Antigen/metabolism , Gastrointestinal Tract/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/metabolism , Tumor Microenvironment , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Animals , B7-H1 Antigen/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Evasion/drug effects , Immune Tolerance/drug effects , Lipopolysaccharides , Lymphocytes, Tumor-Infiltrating/drug effects , Male , Mice, Inbred BALB C , Models, Biological , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
Nat Commun ; 12(1): 5505, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535668

ABSTRACT

Kinase inhibitors suppress the growth of oncogene driven cancer but also enforce the selection of treatment resistant cells that are thought to promote tumor relapse in patients. Here, we report transcriptomic and functional genomics analyses of cells and tumors within their microenvironment across different genotypes that persist during kinase inhibitor treatment. We uncover a conserved, MAPK/IRF1-mediated inflammatory response in tumors that undergo stemness- and senescence-associated reprogramming. In these tumor cells, activation of the innate immunity sensor RIG-I via its agonist IVT4, triggers an interferon and a pro-apoptotic response that synergize with concomitant kinase inhibition. In humanized lung cancer xenografts and a syngeneic Egfr-driven lung cancer model these effects translate into reduction of exhausted CD8+ T cells and robust tumor shrinkage. Overall, the mechanistic understanding of MAPK/IRF1-mediated intratumoral reprogramming may ultimately prolong the efficacy of targeted drugs in genetically defined cancer patients.


Subject(s)
DEAD Box Protein 58/metabolism , Immunity, Innate , Inflammation/pathology , MAP Kinase Signaling System , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cytokines/metabolism , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Evasion/drug effects , Immunity, Innate/drug effects , Interferon Regulatory Factor-1/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Oncogenes , Signal Transduction/drug effects
5.
Nucleic Acids Res ; 49(16): 9389-9403, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34387695

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is a key DNA sensor that detects aberrant cytosolic DNA arising from pathogen invasions or genotoxic stresses. Upon binding to DNA, cGAS is activated and catalyzes the synthesis of cyclic GMP-AMP (cGAMP), which induces potent antimicrobial and antitumor responses. Kaposi sarcoma-associated herpesvirus (KSHV) is a human DNA tumor virus that causes Kaposi sarcoma and several other malignancies. We previously reported that KSHV inhibitor of cGAS (KicGAS) encoded by ORF52, inhibits cGAS enzymatic activity, but the underlying mechanisms remained unclear. To define the inhibitory mechanisms, here we performed in-depth biochemical and functional characterizations of KicGAS, and mapped its functional domains. We found KicGAS self-oligomerizes and binds to double stranded DNA cooperatively. This self-oligomerization is essential for its DNA binding and cGAS inhibition. Interestingly, KicGAS forms liquid droplets upon binding to DNA, which requires collective multivalent interactions with DNA mediated by both structured and disordered domains coordinated through the self-oligomerization of KicGAS. We also observed that KicGAS inhibits the DNA-induced phase separation and activation of cGAS. Our findings reveal a novel mechanism by which DNA viruses target the host protein phase separation for suppression of the host sensing of viral nucleic acids.


Subject(s)
Herpesvirus 8, Human/genetics , Host-Pathogen Interactions/genetics , Nucleotidyltransferases/genetics , Sarcoma, Kaposi/genetics , Cytosol/enzymology , Cytosol/microbiology , DNA Breaks, Double-Stranded/drug effects , DNA Damage/genetics , DNA, Viral/genetics , DNA-Binding Proteins/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Immune Evasion/drug effects , Immunity, Innate/genetics , Nucleotides, Cyclic/genetics , Nucleotidyltransferases/antagonists & inhibitors , Sarcoma, Kaposi/drug therapy , Sarcoma, Kaposi/virology , Viral Proteins/genetics
6.
Oncogene ; 40(31): 5002-5012, 2021 08.
Article in English | MEDLINE | ID: mdl-34175886

ABSTRACT

T cell exhaustion plays critical roles in tumor immune evasion. Novel strategies to suppress immune evasion are in urgent need. We aimed to identify potential compounds to target T cell exhaustion and increase response to immune checkpoint inhibitors (ICIs). Differentially expressed genes (DEGs) were identified between tumors with different immune evasion potential by comparing the transcriptome data. DEGs were then analyzed in the Connectivity Map (CMap) platform to identify potential compounds to increase response to ICIs. Gene set enrichment analysis, LDH release assay, Chromatin immunoprecipitation (ChIP), and Co-IP were performed to explore the potential mechanisms in vitro. Patients derived organoids and humanized xenograft mouse model were utilized to validate the finding ex vivo and in vivo. We identified 25 potential compounds that may play critical roles in regulating tumor immune evasion. We further pinpointed a specific compound, dexamethasone, which shows potent anti-tumor effect in multiple cancer cell lines when cocultured with T cells. Dexamethasone can suppress T cell exhaustion by decreasing the activity of two immune checkpoints simultaneously, including PD-L1 and IDO1. Functional study shows dexamethasone can increase the sensitivity of ICIs in coculture system, 3D organoid model and humanized mouse model. Mechanism study shows dexamethasone mediated transcriptional suppression of PD-L1 and IDO1 depends on the nuclear translocation of GR/STAT3 complex. These findings demonstrate dexamethasone can suppress immune evasion by inducing GR/STAT3 mediated downregulation of PD-L1 and IDO1 pathways.


Subject(s)
B7-H1 Antigen/metabolism , Dexamethasone/pharmacology , Immune Evasion/drug effects , Immunosuppressive Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Databases, Genetic , Disease Models, Animal , Gene Expression Profiling , Heterografts , Humans , Lymphocyte Count , Mice , Models, Biological , Protein Binding , Protein Transport , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Mol Cells ; 44(6): 408-421, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34059561

ABSTRACT

The outbreak of coronavirus disease 2019 (COVID-19) has not only affected human health but also diverted the focus of research and derailed the world economy over the past year. Recently, vaccination against COVID-19 has begun, but further studies on effective therapeutic agents are still needed. The severity of COVID-19 is attributable to several factors such as the dysfunctional host immune response manifested by uncontrolled viral replication, type I interferon suppression, and release of impaired cytokines by the infected resident and recruited cells. Due to the evolving pathophysiology and direct involvement of the host immune system in COVID-19, the use of immune-modulating drugs is still challenging. For the use of immune-modulating drugs in severe COVID-19, it is important to balance the fight between the aggravated immune system and suppression of immune defense against the virus that causes secondary infection. In addition, the interplaying events that occur during virus-host interactions, such as activation of the host immune system, immune evasion mechanism of the virus, and manifestation of different stages of COVID-19, are disjunctive and require thorough streamlining. This review provides an update on the immunotherapeutic interventions implemented to combat COVID-19 along with the understanding of molecular aspects of the immune evasion of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which may provide opportunities to develop more effective and promising therapeutics.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/therapy , Immune Evasion/drug effects , Immunologic Factors/therapeutic use , Virus Replication/drug effects , COVID-19/immunology , COVID-19/pathology , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , Clinical Trials as Topic , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dexamethasone/therapeutic use , Drug Combinations , Humans , Immunity, Innate/drug effects , Immunization, Passive/methods , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Peptides/therapeutic use , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Virus Replication/immunology , COVID-19 Serotherapy
8.
Arch Toxicol ; 95(6): 1899-1915, 2021 06.
Article in English | MEDLINE | ID: mdl-33765170

ABSTRACT

T-2 toxin and deoxynivalenol (DON) are type A and B trichothecenes, respectively. They widely occur as pollutants in food and crops and cause a series of toxicities, including immunotoxicity, hepatotoxicity, and neurotoxicity. Oxidative stress is the primary mechanistic basis of these toxic effects. Increasing amounts of evidence have shown that mitochondria are significant targets of apoptosis caused by T-2 toxin- and DON-induced oxidative stress via regulation of Bax/B-cell lymphoma-2 and caspase-3/caspase-9 signaling. DNA methylation and autophagy are involved in oxidative stress related to apoptosis, and hypoxia and immune evasion are related to oxidative stress in this context. Hypoxia induces oxidative stress by stimulating mitochondrial reactive oxygen species production and regulates the expression of cytokines, such as interleukin-1ß and tumor necrosis factor-α. Programmed cell death-ligand 1 is upregulated by these cytokines and by hypoxia-inducible factor-1, which allows it to bind to programmed cell death-1 to enable escape of immune cell surveillance and achievement of immune evasion. This review concentrates on novel findings regarding the oxidative stress mechanisms of the trichothecenes T-2 toxin and DON. Importantly, we discuss the new evidence regarding the connection of hypoxia and immune evasion with oxidative stress in this context. Finally, the trinity of hypoxia, oxidative stress and immune evasion is highlighted. This work will be conducive to an improved understanding of the oxidative stress caused by trichothecene mycotoxins.


Subject(s)
Oxidative Stress/drug effects , T-2 Toxin/toxicity , Trichothecenes/toxicity , Animals , Apoptosis/drug effects , Humans , Hypoxia/chemically induced , Immune Evasion/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Reactive Oxygen Species/metabolism
9.
Nature ; 592(7853): 277-282, 2021 04.
Article in English | MEDLINE | ID: mdl-33545711

ABSTRACT

The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical for virus infection through the engagement of the human ACE2 protein1 and is a major antibody target. Here we show that chronic infection with SARS-CoV-2 leads to viral evolution and reduced sensitivity to neutralizing antibodies in an immunosuppressed individual treated with convalescent plasma, by generating whole-genome ultra-deep sequences for 23 time points that span 101 days and using in vitro techniques to characterize the mutations revealed by sequencing. There was little change in the overall structure of the viral population after two courses of remdesivir during the first 57 days. However, after convalescent plasma therapy, we observed large, dynamic shifts in the viral population, with the emergence of a dominant viral strain that contained a substitution (D796H) in the S2 subunit and a deletion (ΔH69/ΔV70) in the S1 N-terminal domain of the spike protein. As passively transferred serum antibodies diminished, viruses with the escape genotype were reduced in frequency, before returning during a final, unsuccessful course of convalescent plasma treatment. In vitro, the spike double mutant bearing both ΔH69/ΔV70 and D796H conferred modestly decreased sensitivity to convalescent plasma, while maintaining infectivity levels that were similar to the wild-type virus.The spike substitution mutant D796H appeared to be the main contributor to the decreased susceptibility to neutralizing antibodies, but this mutation resulted in an infectivity defect. The spike deletion mutant ΔH69/ΔV70 had a twofold higher level of infectivity than wild-type SARS-CoV-2, possibly compensating for the reduced infectivity of the D796H mutation. These data reveal strong selection on SARS-CoV-2 during convalescent plasma therapy, which is associated with the emergence of viral variants that show evidence of reduced susceptibility to neutralizing antibodies in immunosuppressed individuals.


Subject(s)
COVID-19 Drug Treatment , COVID-19/therapy , COVID-19/virology , Evolution, Molecular , Mutagenesis/drug effects , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Aged , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , Chronic Disease , Genome, Viral/drug effects , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing , Humans , Immune Evasion/drug effects , Immune Evasion/genetics , Immune Evasion/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunization, Passive , Immunosuppression Therapy , Male , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/immunology , Mutation , Phylogeny , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Time Factors , Viral Load/drug effects , Virus Shedding , COVID-19 Serotherapy
10.
Int J Nanomedicine ; 16: 1175-1187, 2021.
Article in English | MEDLINE | ID: mdl-33623381

ABSTRACT

PURPOSE: Although the neutrophil membrane (NM)-based nanoparticulate delivery system has exhibited rapid advances in tumor targeting stemmed from the inherited instinct, the antitumor effect requires further improvement due to inefficient cellular internalization in the absence of specific interactions between NM-coated nanoparticles and tumor cells. METHODS: Herein, we fabricated drug-paclitaxel loaded NM camouflaging nanoparticles (TNM-PN) modified with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), favorable for the cellular internalization. RESULTS: The results showed that TNM-PN exerted a significant cytotoxicity to tumor cells by TRAIL-mediated endocytosis and strong adhesion to inflamed endothelial cells in vitro. Due to TRAIL modification as well as the adhesive interactions between neutrophil and inflamed tumor vascular endothelial cells, tumors in TNM-PN group exhibited almost 2-fold higher fluorescence intensities than that of NM camouflaging nanoparticles and 3-fold higher than that of bare nanoparticles, respectively. Significant tumor inhibition and survival rates of mice were achieved in TNM-PN group as a consequence of prolonged blood circulations to 48 h and preferential tumor accumulations, which was ascribed to targeting adhesion originated from NM to immune evasion and subsequent excellent cellular internalization. CONCLUSION: The research unveiled a novel strategy of amplifying cellular internalization based on NM coating nanotechnology to boost antitumor efficacy.


Subject(s)
Cell Membrane/metabolism , Drug Delivery Systems , Nanoparticles/chemistry , Nanotechnology/methods , Neoplasms/drug therapy , Neutrophils/cytology , Albumins/pharmacology , Albumins/therapeutic use , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Endocytosis/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Immune Evasion/drug effects , Inflammation/pathology , Mice , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Neoplasms/pathology , Neutrophils/drug effects , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RAW 264.7 Cells , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tissue Distribution/drug effects
11.
Cell ; 184(3): 596-614.e14, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508232

ABSTRACT

Checkpoint inhibitors (CPIs) augment adaptive immunity. Systematic pan-tumor analyses may reveal the relative importance of tumor-cell-intrinsic and microenvironmental features underpinning CPI sensitization. Here, we collated whole-exome and transcriptomic data for >1,000 CPI-treated patients across seven tumor types, utilizing standardized bioinformatics workflows and clinical outcome criteria to validate multivariable predictors of CPI sensitization. Clonal tumor mutation burden (TMB) was the strongest predictor of CPI response, followed by total TMB and CXCL9 expression. Subclonal TMB, somatic copy alteration burden, and histocompatibility leukocyte antigen (HLA) evolutionary divergence failed to attain pan-cancer significance. Dinucleotide variants were identified as a source of immunogenic epitopes associated with radical amino acid substitutions and enhanced peptide hydrophobicity/immunogenicity. Copy-number analysis revealed two additional determinants of CPI outcome supported by prior functional evidence: 9q34 (TRAF2) loss associated with response and CCND1 amplification associated with resistance. Finally, single-cell RNA sequencing (RNA-seq) of clonal neoantigen-reactive CD8 tumor-infiltrating lymphocytes (TILs), combined with bulk RNA-seq analysis of CPI-responding tumors, identified CCR5 and CXCL13 as T-cell-intrinsic markers of CPI sensitivity.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , Neoplasms/immunology , T-Lymphocytes/immunology , Biomarkers, Tumor/metabolism , CD8 Antigens/metabolism , Chemokine CXCL13/metabolism , Chromosomes, Human, Pair 9/genetics , Cohort Studies , Cyclin D1/genetics , DNA Copy Number Variations/genetics , Exome/genetics , Gene Amplification , Humans , Immune Evasion/drug effects , Multivariate Analysis , Mutation/genetics , Neoplasms/pathology , Polymorphism, Single Nucleotide/genetics , Receptors, CCR5/metabolism , T-Lymphocytes/drug effects , Tumor Burden/genetics
12.
J Exp Clin Cancer Res ; 40(1): 4, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33390170

ABSTRACT

BACKGROUND: Progranulin (PGRN), as a multifunctional growth factor, is overexpressed in multiple tumors, but the role of PGRN on tumor immunity is still unclear. Here, we studied the effect of PGRN on breast cancer tumor immunity and its possible molecular mechanism. METHODS: The changes of macrophage phenotypes after PGRN treatment were detected by western blot, quantitative polymerase chain reaction (PCR) and flow cytometry. Western blot was used to study the signal molecular mechanism of PGRN regulating this process. The number and localization of immune cells in Wild-type (WT) and PGRN-/- breast cancer tissues were analyzed by immunohistochemical staining and immunofluorescence techniques. The activation and proliferation of CD8+ T cells were measured by flow cytometry. RESULTS: After being treated with PGRN, the expressions of M2 markers and programmed death ligand 1 (PD-L1) on macrophages increased significantly. Signal transducer and activator of transcription 3 (STAT3) signaling pathway inhibitor Stattic significantly inhibited the expression of PD-L1 and M2 related markers induced by PGRN. In WT group, CD8 were co-localized with macrophages and PD-L1, but not tumor cells. The number of immune cells in PGRN-/- breast cancer tissue increased, and their infiltration into tumor parenchyma was also enhanced. Moreover, in the co-culture system, WT peritoneal macrophages not only reduced the ratio of activated CD8+ T cells but also reduced the proportion of proliferating CD8+ T cells. The addition of programmed death receptor 1 (PD-1) and PD-L1 neutralizing antibodies effectively reversed this effect and restored the immune function of CD8+ T cells. CONCLUSION: These results demonstrate that PGRN promotes M2 polarization and PD-L1 expression by activating the STAT3 signaling pathway. Furthermore, through PD-1/PD-L1 interaction, PGRN can promote the breast tumor immune escape. Our research may provide new ideas and targets for clinical breast cancer immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Breast Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/drug effects , Macrophages/drug effects , Progranulins/pharmacology , Tumor-Associated Macrophages/metabolism , Animals , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Humans , Immune Evasion/drug effects , Macrophages/immunology , Mice , Up-Regulation/drug effects
13.
Front Immunol ; 12: 789145, 2021.
Article in English | MEDLINE | ID: mdl-34975884

ABSTRACT

Chagas' disease is a zoonotic parasitic ailment now affecting more than 6 million people, mainly in Latin America. Its agent, the protozoan Trypanosoma cruzi, is primarily transmitted by endemic hematophagous triatomine insects. Transplacental transmission is also important and a main source for the emerging global expansion of this disease. In the host, the parasite undergoes intra (amastigotes) and extracellular infective (trypomastigotes) stages, both eliciting complex immune responses that, in about 70% of the cases, culminate in permanent immunity, concomitant with the asymptomatic presence of the parasite. The remaining 30% of those infected individuals will develop a syndrome, with variable pathological effects on the circulatory, nervous, and digestive systems. Herein, we review an important number of T. cruzi molecules, mainly located on its surface, that have been characterized as immunogenic and protective in various experimental setups. We also discuss a variety of parasite strategies to evade the complement system - mediated immune responses. Within this context, we also discuss the capacity of the T. cruzi infective trypomastigote to translocate the ER-resident chaperone calreticulin to its surface as a key evasive strategy. Herein, it is described that T. cruzi calreticulin inhibits the initial stages of activation of the host complement system, with obvious benefits for the parasite. Finally, we speculate on the possibility to experimentally intervene in the interaction of calreticulin and other T. cruzi molecules that interact with the complement system; thus resulting in significant inhibition of T. cruzi infectivity.


Subject(s)
Antiprotozoal Agents/pharmacology , Chagas Disease/drug therapy , Host-Parasite Interactions/immunology , Immune Evasion/drug effects , Trypanosoma cruzi/immunology , Antiprotozoal Agents/therapeutic use , Calreticulin/metabolism , Chagas Disease/immunology , Chagas Disease/parasitology , Complement Activation/drug effects , Complement Activation/immunology , Complement System Proteins/metabolism , Humans , Protein Binding/drug effects , Protein Binding/immunology , Protozoan Proteins/metabolism , Trypanosoma cruzi/metabolism
14.
Environ Toxicol Pharmacol ; 81: 103520, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33132153

ABSTRACT

Many diverse strategies allow and facilitate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to evade antiviral innate immune mechanisms. Although the type I interferon (IFN) system has a critical role in restricting the dissemination of viral infection, suppression of IFN receptor signals by SARS-CoV-2 constitutes a checkpoint that plays an important role in the immune escape of the virus. Environmental pollution not only facilitates SARS-CoV-2 infection but also increases infection-associated fatality risk, which arises due to Systemic Aryl hydrocarbon Receptor (AhR) Activation Syndrome. The intracellular accumulation of endogenous kynurenic acid due to overexpression of the indoleamine 2,3-dioxygenase (IDO) by AhR activation induces AhR-interleukin-6 (IL-6)-signal transducers and activators of the transcription 3 (STAT3) signaling pathway. The AhR-IDO1-Kynurenine pathway is an important checkpoint, which leads to fatal consequences in SARS-CoV-2 infection and immune evasion in the context of Treg/Th17 imbalance and cytokine storm.


Subject(s)
COVID-19/immunology , Environmental Pollution/adverse effects , Immune Evasion/immunology , Immunity, Innate/immunology , Inflammation Mediators/immunology , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/metabolism , Humans , Immune Evasion/drug effects , Immunity, Innate/drug effects , Inflammation Mediators/metabolism , Receptors, Aryl Hydrocarbon/immunology , Receptors, Aryl Hydrocarbon/metabolism , COVID-19 Drug Treatment
15.
Int J Nanomedicine ; 15: 10183-10197, 2020.
Article in English | MEDLINE | ID: mdl-33363374

ABSTRACT

PURPOSE: Nanoparticle (NP)-based chemo-photothermal therapy (CPT) has been shown to be a promising non-invasive approach for antitumor treatment. However, NPs must overcome the limitations of opsonization, clearance of the reticuloendothelial system, and ineffective targeting of tumor tissue sites. To solve these problems, stem cell membrane (SCM)-camouflaged polydopamine nanoparticles (PDA@SCM NPs) carrying the hydrophobic anticancer drug 7-ethyl-10-hydroxycamptothecin (SN38) were constructed for CPT of malignant bone tumors. METHODS: We developed umbilical-cord mesenchymal stem cell membrane-coated polydopamine nanoparticles encapsulating SN38 (PDA-SN38@SCM NPs) as an efficient tumor-targeting drug-delivery platform for CPT of malignant bone tumors. We characterized PDA@SCM NPs and evaluated the biocompatibility and anti-phagocytosis properties of PDA@SCM NPs. The antitumor activity of PDA-SN38@SCM NPs was evaluated in MG63 lines and an MG63 xenograft model in mice. RESULTS: Synthesized PDA-SN38@SCM NPs retained an excellent photothermal effect after SN38 loading. The drug release of PDA-SN38@SCM NPs could be triggered by near-infrared irradiation and an acidic stimulus. PDA@SCM NPs exhibited lower nonspecific macrophage uptake, longer retention in blood, and more effective accumulation at tumor sites than that shown by PDA NPs. Confocal laser scanning microscopy (CLSM) and flow cytometry showed that MG63 cells took up more PDA-SN38@SCM NPs than PDA-SN38 NPs. In vitro and in vivo antitumor studies demonstrated the outstanding performance of PDA-SN38@SCM NPs in synergistic CPT for bone tumors. CONCLUSION: PDA-SN38@SCM NPs demonstrated an extraordinary synergistic CPT effect and could be a promising strategy for the treatment of malignant bone tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/therapy , Cell Membrane/metabolism , Indoles/chemistry , Nanoparticles/chemistry , Photothermal Therapy , Polymers/chemistry , Stem Cells/metabolism , Animals , Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor , Drug Liberation , Female , Humans , Immune Evasion/drug effects , Irinotecan/pharmacokinetics , Irinotecan/pharmacology , Irinotecan/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , RAW 264.7 Cells , Tissue Distribution/drug effects
16.
Nat Commun ; 11(1): 6000, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243993

ABSTRACT

Virus infection may induce excessive interferon (IFN) responses that can lead to host tissue injury or even death. ß-arrestin 2 regulates multiple cellular events through the G protein-coupled receptor (GPCR) signaling pathways. Here we demonstrate that ß-arrestin 2 also promotes virus-induced production of IFN-ß and clearance of viruses in macrophages. ß-arrestin 2 interacts with cyclic GMP-AMP synthase (cGAS) and increases the binding of dsDNA to cGAS to enhance cyclic GMP-AMP (cGAMP) production and the downstream stimulator of interferon genes (STING) and innate immune responses. Mechanistically, deacetylation of ß-arrestin 2 at Lys171 facilitates the activation of the cGAS-STING signaling and the production of IFN-ß. In vitro, viral infection induces the degradation of ß-arrestin 2 to facilitate immune evasion, while a ß-blocker, carvedilol, rescues ß-arrestin 2 expression to maintain the antiviral immune response. Our results thus identify a viral immune-evasion pathway via the degradation of ß-arrestin 2, and also hint that carvedilol, approved for treating heart failure, can potentially be repurposed as an antiviral drug candidate.


Subject(s)
Carvedilol/pharmacology , Immune Evasion/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Virus Diseases/immunology , beta-Arrestin 2/metabolism , Animals , Carvedilol/therapeutic use , Disease Models, Animal , Drug Repositioning , HEK293 Cells , Herpesvirus 1, Human/immunology , Humans , Immune Evasion/drug effects , Interferon-beta/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Primary Cell Culture , Proteolysis/drug effects , RAW 264.7 Cells , RNA-Seq , Sendai virus/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Vesiculovirus/immunology , Virus Diseases/drug therapy , Virus Diseases/virology , beta-Arrestin 2/agonists , beta-Arrestin 2/genetics
17.
Int J Mol Med ; 46(5): 1794-1804, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33000204

ABSTRACT

Drug resistance and immune escape of tumor cells severely compromise the treatment efficiency of hepatocellular carcinoma (HCC). Long non­coding RNA KCNQ1 overlapping transcript 1 (lncRNA KCNQ1OT1) has been shown to be involved in drug resistance in several cancers. The aim of the present study was to investigate the role of KCNQ1OT1 in sorafenib resistance and immune escape of HCC cells. Reverse transcription­quantitative PCR analysis, western blotting and immunohistochemistry were performed to detect the expression of KCNQ1OT1, miR­506 and programmed death­ligand­1 (PD­L1). Cell Counting Kit­8 assay, flow cytometry and Transwell assays were used to evaluate IC50 value, cell apoptosis and metastasis. ELISA was performed to detect the secretion of cytokines. Dual­luciferase reporter assay was conducted to verify the targeting relationships between miR­506 and KCNQ1OT1 or PD­L1. KCNQ1OT1 and PD­L1 were found to be upregulated and miR­506 was downregulated in sorafenib­resistant HCC tissues and cells. Furthermore, KCNQ1OT1 knockdown reduced the IC50 value of sorafenib, suppressed cell metastasis and promoted apoptosis in sorafenib­resistant HCC cells. Moreover, KCNQ1OT1 knockdown changed the tumor microenvironment and T­cell apoptosis in a sorafenib­resistant HCC/T­cell co­culture model. In addition, it was demonstrated that KCNQ1OT1 functioned as a competing endogenous RNA of miR­506 and increased PD­L1 expression in sorafenib­resistant HCC cells. miR­506 inhibition abolished the effects of KCNQ1OT1 knockdown on sorafenib sensitivity, tumor growth, the tumor microenvironment and T­cell apoptosis. In conclusion, KCNQ1OT1 knockdown inhibited sorafenib resistance and PD­L1­mediated immune escape by sponging miR­506 in sorafenib­resistant HCC cells.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm , Immune Evasion , Liver Neoplasms/genetics , MicroRNAs/metabolism , Sorafenib/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Evasion/drug effects , Immune Evasion/genetics , MicroRNAs/genetics , Models, Biological , Neoplasm Invasiveness , Potassium Channels, Voltage-Gated/metabolism , Sorafenib/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Microenvironment/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Curr HIV/AIDS Rep ; 17(5): 522-528, 2020 10.
Article in English | MEDLINE | ID: mdl-32875516

ABSTRACT

PURPOSE OF REVIEW: Symptomatic cerebrospinal fluid (CSF) HIV escape defines the presence of neurological disease in combination antiretroviral therapy (cART)-treated persons due to HIV replication in CSF despite systemic suppression or to higher viral replication in CSF than in plasma. The aim was to search for cases of recurrent symptomatic CSF escape and to define their characteristics. RECENT FINDINGS: By review of the literature, we identified symptomatic CSF escape relapses in three patients who had shown clinical remission of a first escape episode following cART optimization. By examination of our cohort of 21 patients with symptomatic CSF escape, we identified five additional patients. In the latter, viral escape relapsed over a median follow-up of 108 months because of low adherence or upon treatment simplification of a previously optimized regimen. cART reoptimization based on resistance profile and potential drug neuropenetration and efficacy led to relapse resolution with no further episodes after a median follow-up of 50 months from relapse. The observation that CSF escape may relapse highlights the importance of long-term neuro-suppressive regimens after a first episode and supports the role of the brain as a reservoir for HIV.


Subject(s)
Anti-HIV Agents/therapeutic use , Cerebrospinal Fluid/virology , HIV Infections/cerebrospinal fluid , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/immunology , Adult , Chronic Disease , Female , HIV Infections/pathology , Humans , Immune Evasion/drug effects , Immune Evasion/immunology , Male , RNA, Viral/blood , Recurrence , Viral Load/drug effects , Virus Replication
19.
Rev Med Virol ; 30(5): e2123, 2020 09.
Article in English | MEDLINE | ID: mdl-32648313

ABSTRACT

The outbreak of coronavirus disease 2019 (COVID-19) and pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a major concern globally. As of 14 April 2020, more than 1.9 million COVID-19 cases have been reported in 185 countries. Some patients with COVID-19 develop severe clinical manifestations, while others show mild symptoms, suggesting that dysregulation of the host immune response contributes to disease progression and severity. In this review, we have summarized and discussed recent immunological studies focusing on the response of the host immune system and the immunopathology of SARS-CoV-2 infection as well as immunotherapeutic strategies for COVID-19. Immune evasion by SARS-CoV-2, functional exhaustion of lymphocytes, and cytokine storm have been discussed as part of immunopathology mechanisms in SARS-CoV-2 infection. Some potential immunotherapeutic strategies to control the progression of COVID-19, such as passive antibody therapy and use of interferon αß and IL-6 receptor (IL-6R) inhibitor, have also been discussed. This may help us to understand the immune status of patients with COVID-19, particularly those with severe clinical presentation, and form a basis for further immunotherapeutic investigations.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antiviral Agents/therapeutic use , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Immune Evasion/drug effects , Immunologic Factors/therapeutic use , Interferon Type I/therapeutic use , Pneumonia, Viral/drug therapy , Betacoronavirus/drug effects , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/therapy , Coronavirus Infections/virology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology , Disease Progression , Gene Expression Regulation , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Immunization, Passive/methods , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/virology , Molecular Targeted Therapy/methods , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , SARS-CoV-2 , Severity of Illness Index , Signal Transduction , COVID-19 Serotherapy
20.
Cancer Cell ; 38(1): 79-96.e11, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32531268

ABSTRACT

Fat mass and obesity-associated protein (FTO), an RNA N6-methyladenosine (m6A) demethylase, plays oncogenic roles in various cancers, presenting an opportunity for the development of effective targeted therapeutics. Here, we report two potent small-molecule FTO inhibitors that exhibit strong anti-tumor effects in multiple types of cancers. We show that genetic depletion and pharmacological inhibition of FTO dramatically attenuate leukemia stem/initiating cell self-renewal and reprogram immune response by suppressing expression of immune checkpoint genes, especially LILRB4. FTO inhibition sensitizes leukemia cells to T cell cytotoxicity and overcomes hypomethylating agent-induced immune evasion. Our study demonstrates that FTO plays critical roles in cancer stem cell self-renewal and immune evasion and highlights the broad potential of targeting FTO for cancer therapy.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/antagonists & inhibitors , Cell Self Renewal/drug effects , Enzyme Inhibitors/pharmacology , Immune Evasion/drug effects , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/chemistry , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Anthracenes/chemistry , Anthracenes/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Evasion/genetics , Leukemia/genetics , Leukemia/pathology , Leukemia/prevention & control , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Structure , Protein Binding/drug effects , Protein Domains , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL