Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 8.443
1.
Signal Transduct Target Ther ; 9(1): 142, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38825657

Radiotherapy combined with immune checkpoint blockade holds great promise for synergistic antitumor efficacy. Targeted radionuclide therapy delivers radiation directly to tumor sites. LNC1004 is a fibroblast activation protein (FAP)-targeting radiopharmaceutical, conjugated with the albumin binder Evans Blue, which has demonstrated enhanced tumor uptake and retention in previous preclinical and clinical studies. Herein, we demonstrate that 68Ga/177Lu-labeled LNC1004 exhibits increased uptake and prolonged retention in MC38/NIH3T3-FAP and CT26/NIH3T3-FAP tumor xenografts. Radionuclide therapy with 177Lu-LNC1004 induced a transient upregulation of PD-L1 expression in tumor cells. The combination of 177Lu-LNC1004 and anti-PD-L1 immunotherapy led to complete eradication of all tumors in MC38/NIH3T3-FAP tumor-bearing mice, with mice showing 100% tumor rejection upon rechallenge. Immunohistochemistry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing revealed that combination therapy reprogrammed the tumor microenvironment in mice to foster antitumor immunity by suppressing malignant progression and increasing cell-to-cell communication, CD8+ T-cell activation and expansion, M1 macrophage counts, antitumor activity of neutrophils, and T-cell receptor diversity. A preliminary clinical study demonstrated that 177Lu-LNC1004 was well-tolerated and effective in patients with refractory cancers. Further, scRNA-seq of peripheral blood mononuclear cells underscored the importance of addressing immune evasion through immune checkpoint blockade treatment. This was emphasized by the observed increase in antigen processing and presentation juxtaposed with T cell inactivation. In conclusion, our data supported the efficacy of immunotherapy combined with 177Lu-LNC1004 for cancer patients with FAP-positive tumors.


Immune Checkpoint Inhibitors , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Endopeptidases/genetics , NIH 3T3 Cells , Radiopharmaceuticals/therapeutic use , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Xenograft Model Antitumor Assays , Immunotherapy , Gelatinases/genetics , Gelatinases/immunology , Lutetium/pharmacology , Cell Line, Tumor
2.
Cell Commun Signal ; 22(1): 305, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38831299

As a major component of innate immunity and a positive regulator of interferons, the Stimulator of interferon gene (STING) has an immunotherapy potential to govern a variety of infectious diseases. Despite the recent advances regarding vaccines against COVID-19, nontoxic novel adjuvants with the potential to enhance vaccine efficacy are urgently desired. In this connection, it has been well-documented that STING agonists are applied to combat COVID-19. This approach is of major significance for boosting immune responses most likely through an autophagy-dependent manner in susceptible individuals against infection induced by severe acute respiratory syndrome Coronavirus (SARS­CoV­2). Given that STING agonists exert substantial immunomodulatory impacts under a wide array of pathologic conditions, these agents could be considered novel adjuvants for enhancing immunogenicity against the SARS-related coronavirus. Here, we intend to discuss the recent advances in STING agonists' recruitment to boost innate immune responses upon vaccination against SARS-related coronavirus infections. In light of the primordial role of autophagy modulation, the potential of being an antiviral vaccine adjuvant was also explored.


Autophagy , COVID-19 , Membrane Proteins , SARS-CoV-2 , Autophagy/immunology , Autophagy/drug effects , Humans , Membrane Proteins/immunology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , Animals , COVID-19 Vaccines/immunology , Immunity, Innate/drug effects , Adjuvants, Vaccine/therapeutic use , Adjuvants, Vaccine/pharmacology , Adjuvants, Immunologic/pharmacology
3.
Parasites Hosts Dis ; 62(2): 193-204, 2024 May.
Article En | MEDLINE | ID: mdl-38835260

Malaria is a global disease affecting a large portion of the world's population. Although vaccines have recently become available, their efficacies are suboptimal. We generated virus-like particles (VLPs) that expressed either apical membrane antigen 1 (AMA1) or microneme-associated antigen (MIC) of Plasmodium berghei and compared their efficacy in BALB/c mice. We found that immune sera acquired from AMA1 VLP- or MIC VLP-immunized mice specifically interacted with the antigen of choice and the whole P. berghei lysate antigen, indicating that the antibodies were highly parasite-specific. Both VLP vaccines significantly enhanced germinal center B cell frequencies in the inguinal lymph nodes of mice compared with the control, but only the mice that received MIC VLPs showed significantly enhanced CD4+ T cell responses in the blood following P. berghei challenge infection. AMA1 and MIC VLPs significantly suppressed TNF-α and interleukin-10 production but had a negligible effect on interferon-γ. Both VLPs prevented excessive parasitemia buildup in immunized mice, although parasite burden reduction induced by MIC VLPs was slightly more effective than that induced by AMA1. Both VLPs were equally effective at preventing body weight loss. Our findings demonstrated that the MIC VLP was an effective inducer of protection against murine experimental malaria and should be the focus of further development.


Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Malaria , Membrane Proteins , Mice, Inbred BALB C , Plasmodium berghei , Protozoan Proteins , Vaccines, Virus-Like Particle , Animals , Plasmodium berghei/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Malaria/immunology , Membrane Proteins/immunology , Mice , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Female , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Parasitemia/immunology , Parasitemia/prevention & control , CD4-Positive T-Lymphocytes/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism
4.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200225, 2024 Jul.
Article En | MEDLINE | ID: mdl-38838283

BACKGROUND AND OBJECTIVES: Patients with ongoing seizures are usually not allowed to drive. The prognosis for seizure freedom is favorable in patients with autoimmune encephalitis (AIE) with antibodies against NMDA receptor (NMDAR), leucine-rich glioma-inactivated 1 (LGI1), contactin-associated protein-like 2 (CASPR2), and the gamma-aminobutyric-acid B receptor (GABABR). We hypothesized that after a seizure-free period of 3 months, patients with AIE have a seizure recurrence risk of <20% during the subsequent 12 months. This would render them eligible for noncommercial driving according to driving regulations in several countries. METHODS: This retrospective multicenter cohort study analyzed follow-up data from patients aged 15 years or older with seizures resulting from NMDAR-, LGI1-, CASPR2-, or GABABR-AIE, who had been seizure-free for ≥3 months. We used Kaplan-Meier (KM) estimates for the seizure recurrence risk at 12 months for each antibody group and tested for the effects of potential covariates with regression models. RESULTS: We included 383 patients with NMDAR-, 440 with LGI1-, 114 with CASPR2-, and 44 with GABABR-AIE from 14 international centers. After being seizure-free for 3 months after an initial seizure period, we calculated the probability of remaining seizure-free for another 12 months (KM estimate) as 0.89 (95% confidence interval [CI] 0.85-0.92) for NMDAR, 0.84 (CI 0.80-0.88) for LGI1, 0.82 (CI 0.75-0.90) for CASPR2, and 0.76 (CI 0.62-0.93) for GABABR. DISCUSSION: Taking a <20% recurrence risk within 12 months as sufficient, patients with NMDAR-AIE and LGI1-AIE could be considered eligible for noncommercial driving after having been seizure-free for 3 months.


Autoantibodies , Encephalitis , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Nerve Tissue Proteins , Receptors, GABA-B , Recurrence , Humans , Female , Male , Adult , Intracellular Signaling Peptides and Proteins/immunology , Autoantibodies/blood , Middle Aged , Encephalitis/immunology , Retrospective Studies , Receptors, GABA-B/immunology , Nerve Tissue Proteins/immunology , Young Adult , Membrane Proteins/immunology , Receptors, N-Methyl-D-Aspartate/immunology , Seizures/etiology , Seizures/immunology , Hashimoto Disease/immunology , Hashimoto Disease/blood , Aged , Adolescent , Follow-Up Studies , Proteins/immunology , Cohort Studies
5.
ACS Nano ; 18(19): 12295-12310, 2024 May 14.
Article En | MEDLINE | ID: mdl-38695532

Immune checkpoint blockade (ICB) has brought tremendous clinical progress, but its therapeutic outcome can be limited due to insufficient activation of dendritic cells (DCs) and insufficient infiltration of cytotoxic T lymphocytes (CTLs). Evoking immunogenic cell death (ICD) is one promising strategy to promote DC maturation and elicit T-cell immunity, whereas low levels of ICD induction of solid tumors restrict durable antitumor efficacy. Herein, we report a genetically edited cell membrane-coated cascade nanozyme (gCM@MnAu) for enhanced cancer immunotherapy by inducing ICD and activating the stimulator of the interferon genes (STING) pathway. In the tumor microenvironment (TME), the gCM@MnAu initiates a cascade reaction and generates abundant cytotoxic hydroxyl (•OH), resulting in improved chemodynamic therapy (CDT) and boosted ICD activation. In addition, released Mn2+ during the cascade reaction activates the STING pathway and further promotes the DC maturation. More importantly, activated immunogenicity in the TME significantly improves gCM-mediated PD-1/PD-L1 checkpoint blockade therapy by eliciting systemic antitumor responses. In breast cancer subcutaneous and lung metastasis models, the gCM@MnAu showed synergistically enhanced therapeutic effects and significantly prolonged the survival of mice. This work develops a genetically edited nanozyme-based therapeutic strategy to improve DC-mediated cross-priming of T cells against poorly immunogenic solid tumors.


Immunotherapy , Animals , Mice , Female , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice, Inbred BALB C , Cell Line, Tumor , Immunogenic Cell Death/drug effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Nanoparticles/chemistry
6.
Front Cell Infect Microbiol ; 14: 1375249, 2024.
Article En | MEDLINE | ID: mdl-38808064

Introduction: Diversity in malarial antigens is an immune evasion mechanism that gives malaria parasites an edge over the host. Immune responses against one variant of a polymorphic antigen are usually not fully effective against other variants due to altered epitopes. This study aimed to evaluate diversity in the Plasmodium falciparum antigens apical membrane antigen 1 (PfAMA1) and circumsporozoite protein (PfCSP) from circulating parasites in a malaria-endemic community in southern Ghana and to determine the effects of polymorphisms on antibody response specificity. Methods: The study involved 300 subjects, whose P. falciparum infection status was determined by microscopy and PCR. Diversity within the two antigens was evaluated by msp2 gene typing and molecular gene sequencing, while the host plasma levels of antibodies against PfAMA1, PfCSP, and two synthetic 24mer peptides from the conserved central repeat region of PfCSP, were measured by ELISA. Results: Of the 300 subjects, 171 (57%) had P. falciparum infection, with 165 of the 171 (96.5%) being positive for either or both of the msp2 allelic families. Gene sequencing of DNA from 55 clonally infected samples identified a total of 56 non-synonymous single nucleotide polymorphisms (SNPs) for the Pfama1 gene and these resulted in 44 polymorphic positions, including two novel positions (363 and 365). Sequencing of the Pfcsp gene from 69 clonal DNA samples identified 50 non-synonymous SNPs that resulted in 42 polymorphic positions, with half (21) of these polymorphic positions being novel. Of the measured antibodies, only anti-PfCSP antibodies varied considerably between PCR parasite-positive and parasite-negative persons. Discussion: These data confirm the presence of a considerable amount of unique, previously unreported amino acid changes, especially within PfCSP. Drivers for this diversity in the Pfcsp gene do not immediately seem apparent, as immune pressure will be expected to drive a similar level of diversity in the Pfama1 gene.


Antibodies, Protozoan , Antigens, Protozoan , Malaria, Falciparum , Membrane Proteins , Plasmodium falciparum , Protozoan Proteins , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Ghana , Humans , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Female , Adult , Male , Adolescent , Young Adult , Child , Genetic Variation , Child, Preschool , Middle Aged , Sequence Analysis, DNA , Enzyme-Linked Immunosorbent Assay , Polymerase Chain Reaction , Antigenic Variation , DNA, Protozoan/genetics
7.
Front Immunol ; 15: 1362159, 2024.
Article En | MEDLINE | ID: mdl-38807595

RNA 5-methylcytosine (m5C) methylation plays a crucial role in hepatocellular carcinoma (HCC). As reported, aberrant m5C methylation is closely associated with the progression, therapeutic efficacy, and prognosis of HCC. The innate immune system functions as the primary defense mechanism in the body against pathogenic infections and tumors since it can activate innate immune pathways through pattern recognition receptors to exert anti-infection and anti-tumor effects. Recently, m5C methylation has been demonstrated to affect the activation of innate immune pathways including TLR, cGAS-STING, and RIG-I pathways by modulating RNA function, unveiling new mechanisms underlying the regulation of innate immune responses by tumor cells. However, research on m5C methylation and its interplay with innate immune pathways is still in its infancy. Therefore, this review details the biological significance of RNA m5C methylation in HCC and discusses its potential regulatory relationship with TLR, cGAS-STING, and RIG-I pathways, thereby providing fresh insights into the role of RNA methylation in the innate immune mechanisms and treatment of HCC.


Carcinoma, Hepatocellular , Immunity, Innate , Liver Neoplasms , RNA Methylation , Animals , Humans , 5-Methylcytosine/metabolism , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , RNA/genetics , Signal Transduction/immunology , RNA Methylation/immunology
8.
Sci Rep ; 14(1): 12374, 2024 05 29.
Article En | MEDLINE | ID: mdl-38811642

Circulating tumor cells (CTCs) have gathered attention as a biomarker for carcinomas. However, CTCs in sarcomas have received little attention. In this work, we investigated cell surface proteins and antibody combinations for immunofluorescence detection of sarcoma CTCs. A microfluidic device that combines filtration and immunoaffinity using gangliosides 2 and cell surface vimentin (CSV) antibodies was employed to capture CTCs. For CTC detection, antibodies against cytokeratins 7 and 8 (CK), pan-cytokeratin (panCK), or a combination of panCK and CSV were used. Thirty-nine blood samples were collected from 21 patients of various sarcoma subtypes. In the independent samples study, samples were subjected to one of three antibody combination choices. Significant difference in CTC enumeration was found between CK and panCK + CSV, and between panCK and panCK + CSV. Upon stratification of CK+ samples, those of metastatic disease had a higher CTC number than those of localized disease. In the paired samples study involving cytokeratin-positive sarcoma subtypes, using panCK antibody detected more CTCs than CK. Similarly, for osteosarcoma, using panCK + CSV combination resulted in a higher CTC count than panCK. This study emphasized deliberate selection of cell surface proteins for sarcoma CTC detection and subtype stratification for studying cancers as heterogeneous as sarcomas.


Biomarkers, Tumor , Neoplastic Cells, Circulating , Sarcoma , Humans , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Sarcoma/pathology , Sarcoma/blood , Sarcoma/diagnosis , Sarcoma/metabolism , Biomarkers, Tumor/blood , Female , Male , Membrane Proteins/metabolism , Membrane Proteins/immunology , Keratins/immunology , Keratins/metabolism , Middle Aged , Adult , Vimentin/metabolism , Vimentin/immunology , Aged , Antibodies/immunology , Cell Line, Tumor
9.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Article En | MEDLINE | ID: mdl-38771879

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Bacterial Proteins , Macrophages , Membrane Proteins , Staphylococcal Infections , Staphylococcus aureus , Type VII Secretion Systems , Ubiquitination , Staphylococcus aureus/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Type VII Secretion Systems/metabolism , Type VII Secretion Systems/immunology , Type VII Secretion Systems/genetics , Mice , Immune Evasion , Host-Pathogen Interactions/immunology
10.
Nat Commun ; 15(1): 4153, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755212

Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis.


ADAM Proteins , Cardiovirus Infections , Encephalomyocarditis virus , Immunity, Innate , Interferon Type I , Interferon-Induced Helicase, IFIH1 , Membrane Proteins , Mice, Knockout , Myocarditis , Animals , Encephalomyocarditis virus/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , ADAM Proteins/metabolism , ADAM Proteins/genetics , ADAM Proteins/immunology , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Myocarditis/immunology , Myocarditis/virology , Humans , Mice, Inbred C57BL , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Signal Transduction/immunology , Male , HEK293 Cells
11.
PLoS One ; 19(5): e0302817, 2024.
Article En | MEDLINE | ID: mdl-38743659

BACKGROUND: IgG subclass levels in hemochromatosis are incompletely characterized. METHODS: We characterized IgG subclass levels of referred hemochromatosis probands with HFE p.C282Y/p.C282Y (rs1800562) and human leukocyte antigen (HLA)-A and -B typing/haplotyping and compared them with IgG subclass levels of eight published cohorts of adults unselected for hemochromatosis. RESULTS: There were 157 probands (82 men, 75 women; mean age 49±13 y). Median serum ferritin, mean body mass index (BMI), median IgG4, and median phlebotomy units to achieve iron depletion were significantly higher in men. Diabetes, cirrhosis, and HLA-A*03,-B*44, -A*03,B*07, and -A*01,B*08 prevalences and median absolute lymphocyte counts in men and women did not differ significantly. Mean IgG subclass levels [95% confidence interval] were: IgG1 5.31 g/L [3.04, 9.89]; IgG2 3.56 g/L [1.29, 5.75]; IgG3 0.61 g/L [0.17, 1.40]; and IgG4 0.26 g/L [<0.01, 1.25]. Relative IgG subclasses were 54.5%, 36.6%, 6.3%, and 2.7%, respectively. Median IgG4 was higher in men than women (0.34 g/L [0.01, 1.33] vs. 0.19 g/L [<0.01, 0.75], respectively; p = 0.0006). A correlation matrix with Bonferroni correction revealed the following positive correlations: IgG1 vs. IgG3 (p<0.01); IgG2 vs. IgG3 (p<0.05); and IgG2 vs. IgG4 (p<0.05). There was also a positive correlation of IgG4 vs. male sex (p<0.01). Mean IgG1 was lower and mean IgG2 was higher in probands than seven of eight published adult cohorts unselected for hemochromatosis diagnoses. CONCLUSIONS: Mean IgG subclass levels of hemochromatosis probands were 5.31, 3.56, 0.61, and 0.26 g/L, respectively. Median IgG4 was higher in men than women. There were positive associations of IgG subclass levels. Mean IgG1 may be lower and mean IgG2 may be higher in hemochromatosis probands than adults unselected for hemochromatosis.


Hemochromatosis Protein , Hemochromatosis , Immunoglobulin G , Humans , Male , Hemochromatosis/blood , Hemochromatosis/genetics , Hemochromatosis/immunology , Female , Immunoglobulin G/blood , Middle Aged , Hemochromatosis Protein/genetics , Adult , Aged , Membrane Proteins/immunology , Membrane Proteins/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology
12.
Viruses ; 16(5)2024 05 06.
Article En | MEDLINE | ID: mdl-38793616

Interferons (IFNs) are antiviral cytokines that defend against viral infections by inducing the expression of interferon-stimulated genes (ISGs). Interferon-inducible transmembrane proteins (IFITMs) 1, 2, and 3 are crucial ISG products and members of the CD225 protein family. Compelling evidence shows that IFITMs restrict the infection of many unrelated viruses by inhibiting the virus-cell membrane fusion at the virus entry step via the modulation of lipid composition and membrane properties. Meanwhile, viruses can evade IFITMs' restrictions by either directly interacting with IFITMs via viral glycoproteins or by altering the native entry pathway. At the same time, cumulative evidence suggests context-dependent and multifaceted roles of IFITMs in modulating virus infections and cell signaling. Here, we review the diverse antiviral mechanisms of IFITMs, the viral antagonizing strategies, and the regulation of IFITM activity in host cells. The mechanisms behind the antiviral activity of IFITMs could aid the development of broad-spectrum antivirals and enhance preparedness for future pandemics.


Interferons , Membrane Proteins , Virus Internalization , Humans , Membrane Proteins/metabolism , Membrane Proteins/immunology , Interferons/immunology , Interferons/metabolism , Virus Internalization/drug effects , Antiviral Agents/pharmacology , Immune Evasion , Animals , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Viruses/drug effects , Host-Pathogen Interactions/immunology , Signal Transduction , Antigens, Differentiation/metabolism , Antigens, Differentiation/immunology
13.
Int J Biol Macromol ; 269(Pt 2): 132104, 2024 Jun.
Article En | MEDLINE | ID: mdl-38719016

Stimulator of interferon genes (STING), as an imperative adaptor protein in innate immune, responds to nucleic acid from invading pathogens to build antiviral responses in host cells. Aberrant activation of STING may trigger tissue damage and autoimmune diseases. Given the decisive role in initiating innate immune response, the activity of STING is intricately governed by several posttranslational modifications, including phosphorylation and ubiquitination. Here, we cloned and characterized a novel RNF122 homolog from common carp (named CcRNF122L). Expression analysis disclosed that the expression of CcRNF122L is up-regulated under spring viremia of carp virus (SVCV) stimulation in vivo and in vitro. Overexpression of CcRNF122L hampers SVCV- or poly(I:C)-mediated the expression of IFN-1 and ISGs in a dose-dependent way. Mechanistically, CcRNF122L interacts with STING and promotes the polyubiquitylation of STING. This polyubiquitylation event inhibits the aggregation of STING and the subsequent recruitment of TBK1 and IRF3 to the signaling complex. Additionally, the deletion of the TM domain abolishes the negative regulatory function of CcRNF122L. Collectively, our discoveries unveil a mechanism that governs the STING function and the precise adjustment of the innate immune response in teleost.


Carps , Fish Proteins , Immunity, Innate , Membrane Proteins , Rhabdoviridae , Animals , Carps/immunology , Carps/genetics , Carps/virology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Rhabdoviridae/physiology , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Fish Diseases/immunology , Fish Diseases/virology , Rhabdoviridae Infections/immunology , Signal Transduction
14.
J Control Release ; 370: 691-706, 2024 Jun.
Article En | MEDLINE | ID: mdl-38723671

Vaccination is essential for preventing and controlling infectious diseases, along with reducing mortality. Developing safe and versatile adjuvants to enhance humoral and cellular immune responses to vaccines remains a key challenge in vaccine development. Here, we designed hierarchical mesoporous MOF-801 (HM801) using a Cocamidopropyl betaine (CAPB) and a Pluronics F127 in an aqueous phase system. Meanwhile, we synthesized a novel SARS-CoV-2 nanovaccine (R@M@HM801) with a high loading capacity for both the STING agonist (MSA-2) and the Delta receptor binding domain (Delta-RBD) antigen. R@M@HM801 enhanced MSA-2 and RBD utilization and effectively co-delivered MSA-2 and RBD antigens to antigen-presenting cells in the draining lymph nodes, thereby promoting the activation of both T and B cells. Lymphocyte single-cell analysis showed that R@M@HM801 stimulated robust CD11b+CD4+ T cells, CXCR5+CD4+ T follicular helper (Tfh), and durable CD4+CD44+CD62L-, CD8+CD44+CD62L- effector memory T cell (TEM) immune responses, and promoted the proliferative activation of CD26+ B cells in vivo. Meanwhile, R@M@HM801 induced stronger specific antibodies and neutralization of pseudovirus against Delta compared to the RBD + MAS-2 and RBD + MAS-2 + Alum vaccines. Our study demonstrated the efficacy of a hierarchical mesoporous HM801 and its potential immune activation mechanism in enhancing adaptive immune responses against viruses and other diseases.


Adjuvants, Immunologic , Immunity, Cellular , Immunity, Humoral , Membrane Proteins , Metal-Organic Frameworks , Animals , Immunity, Humoral/drug effects , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Immunity, Cellular/drug effects , Membrane Proteins/immunology , Mice , Metal-Organic Frameworks/chemistry , Female , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Mice, Inbred BALB C , Porosity , Mice, Inbred C57BL , B-Lymphocytes/immunology , B-Lymphocytes/drug effects
15.
J Immunol ; 212(12): 1945-1957, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38700419

The cytosolic detection of pathogen-derived nucleic acids has evolved as an essential strategy for host innate immune defense in mammals. One crucial component in this process is the stimulator of IFN genes (STING), which acts as a vital signaling adaptor, connecting the cytosolic detection of DNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) to the downstream type I IFN signaling pathway. However, this process remains elusive in invertebrates. In this study, we present evidence demonstrating that STING, an ortholog found in a marine invertebrate (shrimp) called Litopenaeus vannamei, can directly detect DNA and initiate an IFN-like antiviral response. Unlike its homologs in other eukaryotic organisms, which exclusively function as sensors for cyclic dinucleotides, shrimp STING has the ability to bind to both double-stranded DNA and cyclic dinucleotides, including 2'3'-cGAMP. In vivo, shrimp STING can directly sense DNA nucleic acids from an infected virus, accelerate IFN regulatory factor dimerization and nuclear translocation, induce the expression of an IFN functional analog protein (Vago4), and finally establish an antiviral state. Taken together, our findings unveil a novel double-stranded DNA-STING-IKKε-IRF-Vago antiviral axis in an arthropod, providing valuable insights into the functional origins of DNA-sensing pathways in evolution.


Membrane Proteins , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Penaeidae/immunology , Penaeidae/virology , Immunity, Innate/immunology , Signal Transduction/immunology , Interferons/metabolism , Interferons/immunology , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/immunology
16.
Front Immunol ; 15: 1380220, 2024.
Article En | MEDLINE | ID: mdl-38799458

African swine fever (ASF) is an acute hemorrhagic and devastating infectious disease affecting domestic pigs and wild boars. It is caused by the African swine fever virus (ASFV), which is characterized by genetic diversity and sophisticated immune evasion strategies. To facilitate infection, ASFV encodes multiple proteins to antagonize host innate immune responses, thereby contributing to viral virulence and pathogenicity. The molecular mechanisms employed by ASFV-encoded proteins to modulate host antiviral responses have not been comprehensively elucidated. In this study, it was observed that the ASFV MGF505-6R protein, a member of the multigene family 505 (MGF505), effectively suppressed the activation of the interferon-beta (IFN-ß) promoter, leading to reduced mRNA levels of antiviral genes. Additional evidence has revealed that MGF505-6R antagonizes the cGAS-STING signaling pathway by interacting with the stimulator of interferon genes (STING) for degradation in the autophagy-lysosomal pathway. The domain mapping revealed that the N-terminal region (1-260aa) of MGF505-6R is the primary domain responsible for interacting with STING, while the CTT domain of STING is crucial for its interaction with MGF505-6R. Furthermore, MGF505-6R also inhibits the activation of STING by reducing the K63-linked polyubiquitination of STING, leading to the disruption of STING oligomerization and TANK binding kinase 1 (TBK1) recruitment, thereby impairing the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3). Collectively, our study elucidates a novel strategy developed by ASFV MGF505-6R to counteract host innate immune responses. This discovery may offer valuable insights for further exploration of ASFV immune evasion mechanisms and antiviral strategies.


African Swine Fever Virus , African Swine Fever , Membrane Proteins , Viral Proteins , Animals , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Swine , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , Viral Proteins/genetics , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/immunology , Signal Transduction , Proteolysis , HEK293 Cells , Host-Pathogen Interactions/immunology , Immune Evasion , Interferon-beta/metabolism , Interferon-beta/immunology , Interferon-beta/genetics
17.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Article En | MEDLINE | ID: mdl-38729159

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Adjuvants, Immunologic , CD8-Positive T-Lymphocytes , Chitin , Membrane Proteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Signal Transduction , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Signal Transduction/drug effects , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Neoplasms/immunology , Neoplasms/therapy
18.
Viruses ; 16(4)2024 04 09.
Article En | MEDLINE | ID: mdl-38675916

DNA oncoviruses represent an intriguing subject due to their involvement in oncogenesis. These viruses have evolved mechanisms to manipulate the host immune response, facilitating their persistence and actively contributing to carcinogenic processes. This paper describes the complex interactions between DNA oncoviruses and the innate immune system, with a particular emphasis on the cGAS-STING pathway. Exploring these interactions highlights that DNA oncoviruses strategically target and subvert this pathway, exploiting its vulnerabilities for their own survival and proliferation within the host. Understanding these interactions lays the foundation for identifying potential therapeutic interventions. Herein, we sought to contribute to the ongoing efforts in advancing our understanding of the innate immune system in oncoviral pathogenesis.


Immune Evasion , Immunity, Innate , Nucleotidyltransferases , Humans , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Signal Transduction , DNA Tumor Viruses/genetics , DNA Tumor Viruses/immunology , Host-Pathogen Interactions/immunology
19.
Microb Cell Fact ; 23(1): 119, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38659027

BACKGROUND: Clostridium spp. has demonstrated therapeutic potential in cancer treatment through intravenous or intratumoral administration. This approach has expanded to include non-pathogenic clostridia for the treatment of various diseases, underscoring the innovative concept of oral-spore vaccination using clostridia. Recent advancements in the field of synthetic biology have significantly enhanced the development of Clostridium-based bio-therapeutics. These advancements are particularly notable in the areas of efficient protein overexpression and secretion, which are crucial for the feasibility of oral vaccination strategies. Here, we present two examples of genetically engineered Clostridium candidates: one as an oral cancer vaccine and the other as an antiviral oral vaccine against SARS-CoV-2. RESULTS: Using five validated promoters and a signal peptide derived from Clostridium sporogenes, a series of full-length NY-ESO-1/CTAG1, a promising cancer vaccine candidate, expression vectors were constructed and transformed into C. sporogenes and Clostridium butyricum. Western blotting analysis confirmed efficient expression and secretion of NY-ESO-1 in clostridia, with specific promoters leading to enhanced detection signals. Additionally, the fusion of a reported bacterial adjuvant to NY-ESO-1 for improved immune recognition led to the cloning difficulties in E. coli. The use of an AUU start codon successfully mitigated potential toxicity issues in E. coli, enabling the secretion of recombinant proteins in C. sporogenes and C. butyricum. We further demonstrate the successful replacement of PyrE loci with high-expression cassettes carrying NY-ESO-1 and adjuvant-fused NY-ESO-1, achieving plasmid-free clostridia capable of secreting the antigens. Lastly, the study successfully extends its multiplex genetic manipulations to engineer clostridia for the secretion of SARS-CoV-2-related Spike_S1 antigens. CONCLUSIONS: This study successfully demonstrated that C. butyricum and C. sporogenes can produce the two recombinant antigen proteins (NY-ESO-1 and SARS-CoV-2-related Spike_S1 antigens) through genetic manipulations, utilizing the AUU start codon. This approach overcomes challenges in cloning difficult proteins in E. coli. These findings underscore the feasibility of harnessing commensal clostridia for antigen protein secretion, emphasizing the applicability of non-canonical translation initiation across diverse species with broad implications for medical or industrial biotechnology.


Clostridium butyricum , Clostridium , Recombinant Proteins , Clostridium butyricum/genetics , Clostridium butyricum/metabolism , Clostridium/genetics , Clostridium/metabolism , Humans , Recombinant Proteins/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Cancer Vaccines/immunology , Cancer Vaccines/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Administration, Oral , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Spores, Bacterial/genetics , Spores, Bacterial/immunology , Vaccination , COVID-19/prevention & control , Genetic Engineering , Escherichia coli/genetics , Escherichia coli/metabolism , Promoter Regions, Genetic
20.
mBio ; 15(5): e0085924, 2024 May 08.
Article En | MEDLINE | ID: mdl-38639536

Visceral leishmaniasis is a deadly infectious disease and is one of the world's major neglected health problems. Because the symptoms of infection are similar to other endemic diseases, accurate diagnosis is crucial for appropriate treatment. Definitive diagnosis using splenic or bone marrow aspirates is highly invasive, and so, serological assays are preferred, including the direct agglutination test (DAT) or rK39 strip test. These tests, however, are either difficult to perform in the field (DAT) or lack specificity in some endemic regions (rK39), making the development of new tests a research priority. The availability of Leishmania spp. genomes presents an opportunity to identify new diagnostic targets. Here, we use genome data and a mammalian protein expression system to create a panel of 93 proteins consisting of the extracellular ectodomains of the Leishmania donovani cell surface and secreted proteins. We use these panel and sera from murine experimental infection models and natural human and canine infections to identify new candidates for serological diagnosis. We observed a concordance between the most immunoreactive antigens in different host species and transmission settings. The antigen encoded by the LdBPK_323600.1 gene can diagnose Leishmania infections with high sensitivity and specificity in patient cohorts from different endemic regions including Bangladesh and Ethiopia. In longitudinal sampling of treated patients, we observed reductions in immunoreactivity to LdBPK_323600.1 suggesting it could be used to diagnose treatment success. In summary, we have identified new antigens that could contribute to improved serological diagnostic tests to help control the impact of this deadly tropical infectious disease. IMPORTANCE: Visceral leishmaniasis is fatal if left untreated with patients often displaying mild and non-specific symptoms during the early stages of infection making accurate diagnosis important. Current methods for diagnosis require highly trained medical staff to perform highly invasive biopsies of the liver or bone marrow which pose risks to the patient. Less invasive molecular tests are available but can suffer from regional variations in their ability to accurately diagnose an infection. To identify new diagnostic markers of visceral leishmaniasis, we produced and tested a panel of 93 proteins identified from the genome of the parasite responsible for this disease. We found that the pattern of host antibody reactivity to these proteins was broadly consistent across naturally acquired infections in both human patients and dogs, as well as experimental rodent infections. We identified a new protein called LdBPK_323600.1 that could accurately diagnose visceral leishmaniasis infections in humans.


Antibodies, Protozoan , Antigens, Protozoan , Leishmania donovani , Leishmaniasis, Visceral , Protozoan Proteins , Serologic Tests , Leishmania donovani/genetics , Leishmania donovani/immunology , Leishmaniasis, Visceral/diagnosis , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Animals , Humans , Mice , Dogs , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Serologic Tests/methods , Biomarkers/blood , Female , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Mice, Inbred BALB C , Membrane Proteins/genetics , Membrane Proteins/immunology , Sensitivity and Specificity , Dog Diseases/diagnosis , Dog Diseases/parasitology
...