Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Adv Sci (Weinh) ; : e2400149, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898748

ABSTRACT

The activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) and its adaptor, stimulator of interferon genes (STING), is known to reprogram the immunosuppressive tumor microenvironment for promoting antitumor immunity. To enhance the efficiency of cGAS-STING pathway activation, macrophage-selective uptake, and programmable cytosolic release are crucial for the delivery of STING agonists. However, existing polymer- or lipid-based delivery systems encounter difficulty in integrating multiple functions meanwhile maintaining precise control and simple procedures. Herein, inspired by cGAS being a natural DNA sensor, a modularized DNA nanodevice agonist (DNDA) is designed that enable macrophage-selective uptake and programmable activation of the cGAS-STING pathway through precise self-assembly. The resulting DNA nanodevice acts as both a nanocarrier and agonist. Upon local administration, it demonstrates the ability of macrophage-selective uptake, endosomal escape, and cytosolic release of the cGAS-recognizing DNA segment, leading to robust activation of the cGAS-STING pathway and enhanced antitumor efficacy. Moreover, DNDA elicits a synergistic therapeutic effect when combined with immune checkpoint blockade. The study broadens the application of DNA nanotechnology as an immune stimulator for cGAS-STING activation.

2.
Cancer Biol Med ; 20(12)2024 02 05.
Article in English | MEDLINE | ID: mdl-38318930

ABSTRACT

Maintenance immunotherapy after concurrent chemoradiotherapy remains the standard therapeutic approach in patients with unresectable locally advanced non-small cell lung cancer (LA-NSCLC). The efficacy of pembrolizumab without chemotherapy in stage IV NSCLC has incited interest in similar approaches for LA-NSCLC. Several recent investigations involving the synergistic potential of immunotherapy combined with radiotherapy (iRT) have generated encouraging results. This review discusses the existing studies and prospective directions of chemotherapy-free iRT strategies in unresectable LA-NSCLC. Although the initial findings of chemotherapy-free iRT strategies have shown promising efficacy, we must consider the methodologic limitations of current studies and the myriad of challenges that accompany the implementation of chemotherapy-free iRT. These challenges include determining the optimal dose and fractionation, precise target volume delineation, and identification of additional suitable patient cohorts. Furthermore, the feasibility of chemotherapy-free iRT as a novel treatment modality for select patients with LA-NSCLC is contingent upon validation through randomized phase III trials.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Prospective Studies , Chemoradiotherapy/methods
3.
Trends Mol Med ; 30(3): 202-203, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38302316

ABSTRACT

Ma et al. recently reported a systematic screening of small-molecule compounds targeting the FG-CC' cleft of T cell immunoglobulin and mucin-containing molecule 3 (TIM-3). They identified a functional Tim-3 inhibitor, ML-T7, that, as a single agent or in combination with anti-PD-1, demonstrated strong antitumor activity in preclinical mouse tumor models, supporting its potential for further clinical translation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Neoplasms , Animals , Humans , Mice , Immunotherapy , Neoplasms/drug therapy
4.
Cell Rep ; 43(2): 113712, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38294903

ABSTRACT

Adoptive cell therapies are emerging forms of immunotherapy that reprogram T cells for enhanced antitumor responses. Although surface programmed cell death-ligand 1 (PD-L1)/programmed cell death protein 1 (PD-1) engagement inhibits antitumor immunity, the role of cell-intrinsic PD-L1 in adoptive T cell therapy remains unknown. Here, we found that intracellular PD-L1 was enriched in tumor-infiltrating CD8+ T cells of cancer patients. PD-L1 ablation promoted antitumor immune responses and the maintenance of an effector-like state of therapeutic CD8+ T cells, while blockade of surface PD-L1 was unable to impact on their expansion and function. Moreover, cell-intrinsic PD-L1 impeded CD8+ T cell activity, which partially relied on mTORC1 signaling. Furthermore, endogenous tumor-reactive CD8+ T cells were motivated by BATF3-driven dendritic cells after adoptive transfer of PD-L1-deficient therapeutic CD8+ T cells. This role of cell-intrinsic PD-L1 in therapeutic CD8+ T cell dysfunction highlights that disrupting cell-intrinsic PD-L1 in CD8+ T cells represents a viable approach to improving T cell-based cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , B7-H1 Antigen , Immunotherapy , Cell- and Tissue-Based Therapy , Membrane Proteins , Neoplasms/therapy
5.
Sci Immunol ; 8(90): eadf4919, 2023 12.
Article in English | MEDLINE | ID: mdl-38039379

ABSTRACT

Mitochondrial antiviral signaling protein (MAVS)-mediated cytosolic RNA sensing plays a central role in tumor immunogenicity. However, the effects of host MAVS signaling on antitumor immunity remain unclear. Here, we demonstrate that the host MAVS pathway supports tumor growth and impairs antitumor immunity, whereas MAVS deficiency in dendritic cells (DCs) promotes tumor-reactive CD8+ T cell responses. Specifically, CD8+ T cell priming capacity was enhanced by MAVS ablation in a type I interferon-independent, but IL-12-dependent, manner. Mechanistically, loss of the RIG-I/MAVS cascade activated the noncanonical NF-κB pathway and in turn induced IL-12 production by DCs. MAVS-restrained IL-12 promoted cross-talk between CD8+ T cells and DCs, which was licensed by IFN-γ. Moreover, ablation of host MAVS sensitized tumors to immunotherapy and attenuated radiation resistance, thereby facilitating the maintenance of effector CD8+ T cells. These findings demonstrate that the host MAVS pathway acts as an immune regulator of DC-driven antitumor immunity and support the development of immunotherapies that antagonize MAVS signaling in DCs.


Subject(s)
Interleukin-12 , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Signal Transduction , Dendritic Cells
6.
Cell Rep ; 42(9): 113108, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37708030

ABSTRACT

Natural killer (NK) cells are cytotoxic innate lymphocytes that eradicate tumor cells. Inducing durable antitumor immune responses by NK cells represents a major priority of cancer immunotherapy. While cytosolic DNA sensing plays an essential role in initiating antitumor immunity, the role of NK cell-intrinsic STING signaling remains unclear. Here, we find that NK cell-intrinsic STING promotes antitumor responses and maintains a reservoir of TCF-1+ NK cells. In contrast, tumor cell-intrinsic cGAS and mtDNA are required for NK cell antitumor activity, indicating that tumor mtDNA recognition by cGAS partially triggers NK cell-intrinsic STING activation. Moreover, addition of cGAMP enables STING activation and type I interferon production in NK cells, thereby supporting the activation of NK cells in vitro. In humans, STING agonism promotes the expansion of TCF-1+ NK cells. This study provides insight into understanding how STING signaling drives NK cell antitumor immunity and the development of NK cell-based cancer immunotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Nucleotidyltransferases/metabolism , DNA, Mitochondrial , Killer Cells, Natural/metabolism , Immunity, Innate
7.
Chem Sci ; 14(22): 5956-5964, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37293644

ABSTRACT

Controllable activation of the innate immune adapter protein - stimulator of interferon genes (STING) pathway is a critical challenge for the clinical development of STING agonists due to the potential "on-target off-tumor" toxicity caused by systematic activation of STING. Herein, we designed and synthesized a photo-caged STING agonist 2 with a tumor cell-targeting carbonic anhydrase inhibitor warhead, which could be readily uncaged by blue light to release the active STING agonist leading to remarkable activation of STING signaling. Furthermore, compound 2 was found to preferentially target tumor cells, stimulate the STING signaling in zebrafish embryo upon photo-uncaging and to induce proliferation of macrophages and upregulation of the mRNA expression of STING as well as its downstream NF-kB and cytokines, thus leading to significant suppression of tumor cell growth in a photo-dependent manner with reduced systemic toxicity. This photo-caged agonist not only provides a powerful tool to precisely trigger STING signalling, but also represents a novel controllable STING activation strategy for safer cancer immunotherapy.

8.
Immunity ; 56(5): 1013-1026.e6, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36944334

ABSTRACT

Sepsis is a dysregulated inflammatory consequence of systemic infection. As a result, excessive platelet activation leads to thrombosis and coagulopathy, but we currently lack sufficient understanding of these processes. Here, using the cecal ligation and puncture (CLP) model of sepsis, we observed septic thrombosis and neutrophil extracellular trap formation (NETosis) within the mouse vasculature by intravital microscopy. STING activation in platelets was a critical driver of sepsis-induced pathology. Platelet-specific STING deficiency suppressed platelet activation and granule secretion, which alleviated sepsis-induced intravascular thrombosis and NETosis in mice. Mechanistically, sepsis-derived cGAMP promoted the binding of STING to STXBP2, the assembly of SNARE complex, granule secretion, and subsequent septic thrombosis, which probably depended on the palmitoylation of STING. We generated a peptide, C-ST5, to block STING binding to STXBP2. Septic mice treated with C-ST5 showed reduced thrombosis. Overall, platelet activation via STING reveals a potential strategy for limiting life-threatening sepsis-mediated coagulopathy.


Subject(s)
Extracellular Traps , Sepsis , Thrombosis , Animals , Mice , Blood Platelets/metabolism , Extracellular Traps/metabolism , Mice, Inbred C57BL , Munc18 Proteins/metabolism , Platelet Activation , Sepsis/metabolism , Thrombosis/metabolism
10.
Cell Death Dis ; 13(7): 640, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869062

ABSTRACT

Our previous studies show that the mitotic phosphorylation of SUMO-specific protease 3 (SENP3) can inhibit its de-SUMOylation activity in G2/M phase of the cell cycle. Inhibition of SENP3 plays a critical role in the correct separation of sister chromatids in mitosis. The mutation of mitotic SENP3 phosphorylation causes chromosome instability and promotes tumorigenesis. In this study, we find that the mutation of mitotic SENP3 phosphorylation in tumor cells can suppress tumor growth in immune-competent mouse model. We further detect an increase of CD8+ T cell infiltration in the tumors, which is essential for the anti-tumor effect in immune-competent mouse model. Moreover, we find that mitotic SENP3 activation increases micronuclei formation, which can activate cGAS signaling-dependent innate immune response. We confirmed that cGAS signaling mediates the mitotic SENP3 activation-induced anti-tumor immunity. We further show that p53 responding to DNA damage activates mitotic SENP3 by inhibiting phosphorylation, and further increases cellular senescence as well as the related innate immune response in tumor cells. Furthermore, TCGA database demonstrates that the SENP3 expression positively correlates with the induction of innate immune response as well as the survival of the p53 mutant pancreatic cancer patients. Together, these data reveal that mitotic SENP3 activation in tumor cells can promote host anti-tumor immune response by coupling with cGAS signaling.


Subject(s)
Neoplasms , Peptide Hydrolases , Animals , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Mice , Neoplasms/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Peptide Hydrolases/metabolism , Sumoylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Immunity ; 55(7): 1268-1283.e9, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35700739

ABSTRACT

The incidence and mortality rates of many non-reproductive human cancers are generally higher in males than in females. However, the immunological mechanism underlying sexual differences in cancers remains elusive. Here, we demonstrated that sex-related differences in tumor burden depended on adaptive immunity. Male CD8+ T cells exhibited impaired effector and stem cell-like properties compared with female CD8+ T cells. Mechanistically, androgen receptor inhibited the activity and stemness of male tumor-infiltrating CD8+ T cells by regulating epigenetic and transcriptional differentiation programs. Castration combined with anti-PD-L1 treatment synergistically restricted tumor growth in male mice. In humans, fewer male CD8+ T cells maintained a stem cell-like memory state compared with female counterparts. Moreover, AR expression correlated with tumor-infiltrating CD8+ T cell exhaustion in cancer patients. Our findings reveal sex-biased CD8+ T cell stemness programs in cancer progression and in the responses to cancer immunotherapy, providing insights into the development of sex-based immunotherapeutic strategies for cancer treatment.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Female , Humans , Immunotherapy , Male , Mice , Neoplasms/therapy , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sex Characteristics , Tumor Microenvironment
12.
J Cell Biol ; 221(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35510944

ABSTRACT

The stimulator of interferon genes (STING) plays a critical role in innate immunity. Emerging evidence suggests that STING is important for DNA or cGAMP-induced non-canonical autophagy, which is independent of a large part of canonical autophagy machineries. Here, we report that, in the absence of STING, energy stress-induced autophagy is upregulated rather than downregulated. Depletion of STING in Drosophila fat cells enhances basal- and starvation-induced autophagic flux. During acute exercise, STING knockout mice show increased autophagy flux, exercise endurance, and altered glucose metabolism. Mechanistically, these observations could be explained by the STING-STX17 interaction. STING physically interacts with STX17, a SNARE that is essential for autophagosome biogenesis and autophagosome-lysosome fusion. Energy crisis and TBK1-mediated phosphorylation both disrupt the STING-STX17 interaction, allow different pools of STX17 to translocate to phagophores and mature autophagosomes, and promote autophagic flux. Taken together, we demonstrate a heretofore unexpected function of STING in energy stress-induced autophagy through spatial regulation of autophagic SNARE STX17.


Subject(s)
Autophagy , Energy Metabolism , Lysosomes , Membrane Proteins , Qa-SNARE Proteins , Animals , Autophagosomes/metabolism , Drosophila , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Physical Conditioning, Animal , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism
13.
Sci Adv ; 7(41): eabf6290, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34613770

ABSTRACT

Necroptosis, a form of regulated necrosis, participates in tumor development and dying cell immunogenicity. However, it remains unclear how tumor cell­intrinsic necroptotic signaling contributes to radiation-induced antitumor immunity. Here, we found that the ZBP1-MLKL necroptotic cascade in irradiated tumor cells was essential for antitumor immunity. ZBP1-dependent activation of MLKL potentiated type I interferon responses following tumor cell irradiation. Mechanistically, the ZBP1-MLKL necroptotic cascade induced cytoplasmic DNA accumulation in irradiated tumor cells and, in turn, autonomously activated cGAS-STING signaling, thus creating a positive feedback loop between those two pathways to drive persistent inflammation. Accordingly, ablation of caspase-8 enhanced STING pathway activation and the antitumor effects of radiation by activating MLKL. These findings reveal that ZBP1-MLKL necroptosis signaling maximized radiation-induced antitumor immunity through mutual interaction with the tumor cell­intrinsic STING pathway. This study provides insight into how radiotherapy bridges tumor cell damage to antitumor immune responses and an alternative strategy to improve radiotherapy.

14.
Nat Commun ; 12(1): 4371, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272364

ABSTRACT

Metabolic programming and mitochondrial dynamics along with T cell differentiation affect T cell fate and memory development; however, how to control metabolic reprogramming and mitochondrial dynamics in T cell memory development is unclear. Here, we provide evidence that the SUMO protease SENP1 promotes T cell memory development via Sirt3 deSUMOylation. SENP1-Sirt3 signalling augments the deacetylase activity of Sirt3, promoting both OXPHOS and mitochondrial fusion. Mechanistically, SENP1 activates Sirt3 deacetylase activity in T cell mitochondria, leading to reduction of the acetylation of mitochondrial metalloprotease YME1L1. Consequently, deacetylation of YME1L1 suppresses its activity on OPA1 cleavage to facilitate mitochondrial fusion, which results in T cell survival and promotes T cell memory development. We also show that the glycolytic intermediate fructose-1,6-bisphosphate (FBP) as a negative regulator suppresses AMPK-mediated activation of the SENP1-Sirt3 axis and reduces memory development. Moreover, glucose limitation reduces FBP production and activates AMPK during T cell memory development. These data show that glucose limitation activates AMPK and the subsequent SENP1-Sirt3 signalling for T cell memory development.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CD8-Positive T-Lymphocytes/immunology , Cysteine Endopeptidases/metabolism , Immunologic Memory , Mitochondria/metabolism , Sirtuin 3/metabolism , T-Lymphocytes/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , Acetylation , Allografts , Animals , Cell Line, Tumor , Cell Survival/genetics , Colonic Neoplasms/immunology , Fructosediphosphates/metabolism , GTP Phosphohydrolases/metabolism , Glucose/deficiency , Immunologic Memory/genetics , Metabolomics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/ultrastructure , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Sirtuin 3/antagonists & inhibitors , Sirtuin 3/genetics , Sumoylation , T-Lymphocytes/immunology
15.
Mol Cell ; 81(5): 940-952.e5, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33434504

ABSTRACT

STING-dependent cytosolic DNA sensing in dendritic cells (DCs) initiates antitumor immune responses, but how STING signaling is metabolically regulated in the tumor microenvironment remains unknown. Here, we show that oxidative stress is required for STING-induced DC antitumor function through a process that directs SUMO-specific protease 3 (SENP3) activity. DC-specific deletion of Senp3 drives tumor progression by blunting STING-dependent type-I interferon (IFN) signaling in DCs and dampening antitumor immune responses. DC-derived reactive oxygen species (ROS) trigger SENP3 accumulation and the SENP3-IFI204 interaction, thereby catalyzing IFI204 deSUMOylation and boosting STING signaling activation in mice. Consistently, SENP3 senses ROS to facilitate STING-dependent DC activity in tissue samples from colorectal cancer patients. Our results reveal that oxidative stress as a metabolic regulator promotes STING-mediated DC antitumor immune responses and highlights SENP3 as an overflow valve for STING signaling induction in the metabolically abnormal tumor microenvironment.


Subject(s)
Colorectal Neoplasms/genetics , Cysteine Endopeptidases/genetics , Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Allografts , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cysteine Endopeptidases/immunology , Dendritic Cells/pathology , Female , HEK293 Cells , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Oxidative Stress , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
16.
Cancer Lett ; 502: 166-179, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33450361

ABSTRACT

The brain is one of the most common metastatic sites in non-small cell lung cancer (NSCLC), which is associated with an extremely poor prognosis. Despite the availability of several therapeutic options, the treatment efficacy remains unsatisfactory for NSCLC brain metastases. Anti-programmed cell death-1 (PD-1) and its ligand (PD-L1) monoclonal antibodies have reshaped therapeutic strategies in advanced NSCLC. Preliminary evidence has shown that anti-PD-(L)1 monotherapy is also effective in NSCLC patients with brain metastases. However, the traditional view asserted that these therapeutic antibodies were incapable of crossing the blood-brain barrier (BBB) with large molecular size, thus most patients with brain metastases were excluded from most studies on anti-PD-(L)1 immunotherapy. Therefore, the efficacy and its mechanisms of action of anti-PD-(L)1 immunotherapy against brain metastases in NSCLC have not been clarified. In this review, we will survey the underlying mechanisms and current clinical advances of anti-PD-(L)1 immunotherapy in the treatment of brain metastases in NSCLC. The trafficking of activated cytotoxic T cells that are mainly derived from the primary tumor and deep cervical lymph nodes is critical for the intracranial response to anti-PD-(L)1 immunotherapy, which is driven by interferon-γ (IFN-γ). Additionally, promising combined strategies with the rationale in the treatment of brain metastases will be presented to provide future directions for clinical study design. Several significant challenges in the preclinical and clinical studies of brain metastases, as well as potential solutions, will also be discussed.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Brain Neoplasms/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Lung Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors
17.
Sci Transl Med ; 12(549)2020 06 24.
Article in English | MEDLINE | ID: mdl-32581136

ABSTRACT

Although cGAS-STING-mediated DNA sensing in tumor cells or phagocytes is central for launching antitumor immunity, the role of intrinsic cGAS-STING activation in T cells remains unknown. Here, we observed that peripheral blood CD8+ T cells from patients with cancer showed remarkably compromised expression of the cGAS-STING cascade. We demonstrated that the cGAS-STING cascade in adoptively transferred CD8+ T cells was essential for antitumor immune responses in the context of T cell therapy in mice. Mechanistically, cell-autonomous cGAS and STING promoted the maintenance of stem cell-like CD8+ T cells, in part, by regulating the transcription factor TCF1 expression. Moreover, autocrine cGAS-STING-mediated type I interferon signaling augmented stem cell-like CD8+ T cell differentiation program mainly by restraining Akt activity. In addition, genomic DNA was selectively enriched in the cytosol of mouse CD8+ T cells upon in vitro and in vivo stimulation. STING agonism enhanced the formation of stem-like central memory CD8+ T cells from patients with cancer and potentiated antitumor responses of CAR-T cell therapy in a xenograft model. These findings advance our understanding of inherent cGAS-STING activation in T cells and provide insight into the development of improved T cell therapy by harnessing the cGAS-STING pathway for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Membrane Proteins , Animals , Cell- and Tissue-Based Therapy , DNA , Humans , Mice , Nucleotidyltransferases
18.
Immunity ; 49(3): 490-503.e4, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30170810

ABSTRACT

The NF-κB pathway plays a crucial role in supporting tumor initiation, progression, and radioresistance of tumor cells. However, the role of the NF-κB pathway in radiation-induced anti-tumor host immunity remains unclear. Here we demonstrated that inhibiting the canonical NF-κB pathway dampened the therapeutic effect of ionizing radiation (IR), whereas non-canonical NF-κB deficiency promoted IR-induced anti-tumor immunity. Mechanistic studies revealed that non-canonical NF-κB signaling in dendritic cells (DCs) was activated by the STING sensor-dependent DNA-sensing pathway. By suppressing recruitment of the transcription factor RelA onto the Ifnb promoter, activation of the non-canonical NF-κB pathway resulted in decreased type I IFN expression. Administration of a specific inhibitor of the non-canonical NF-κB pathway enhanced the anti-tumor effect of IR in murine models. These findings reveal the potentially interactive roles for canonical and non-canonical NF-κB pathways in IR-induced STING-IFN production and provide an alternative strategy to improve cancer radiotherapy.


Subject(s)
Colonic Neoplasms/radiotherapy , Dendritic Cells/immunology , Melanoma/radiotherapy , NF-kappa B/metabolism , Neoplasms, Experimental/radiotherapy , Radiotherapy/methods , Receptors, Pattern Recognition/metabolism , Animals , Colonic Neoplasms/immunology , DNA/immunology , Disease Models, Animal , Humans , Immunity, Cellular , Melanoma/immunology , Melanoma, Experimental , Membrane Proteins/metabolism , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Radiation Tolerance , Radiation, Ionizing , Signal Transduction , Transcription Factor RelA/metabolism , Xenograft Model Antitumor Assays
19.
Nat Commun ; 8(1): 1736, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170400

ABSTRACT

Radiotherapy induces and promotes innate and adaptive immunity in which host STING plays an important role. However, radioresistance in irradiated tumors can also develop, resulting in relapse. Here we report a mechanism by which extrinsic resistance develops after local ablative radiation that relies on the immunosuppressive action of STING. The STING/type I interferon pathway enhances suppressive inflammation in tumors by recruiting myeloid cells in part via the CCR2 pathway. Germ-line knockouts of CCR2 or treatment with an anti-CCR2 antibody results in blockade of radiation-induced MDSC infiltration. Treatment with anti-CCR2 antibody alleviates immunosuppression following activation of the STING pathway, enhancing the anti-tumor effects of STING agonists and radiotherapy. We propose that radiation-induced STING activation is immunosuppressive due to (monocytic) M-MDSC infiltration, which results in tumor radioresistance. Furthermore, the immunosuppressive effects of radiotherapy and STING agonists can be abrogated in humans by a translational strategy involving anti-CCR2 antibody treatment to improve radiotherapy.


Subject(s)
Membrane Proteins/immunology , Myeloid-Derived Suppressor Cells/immunology , Radiation Tolerance/immunology , Adaptive Immunity , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/radiotherapy , Female , Humans , Immunity, Innate , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Signal Transduction
20.
Immunity ; 47(2): 363-373.e5, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801234

ABSTRACT

Inhibition of cytosolic DNA sensing represents a strategy that tumor cells use for immune evasion, but the underlying mechanisms are unclear. Here we have shown that CD47-signal regulatory protein α (SIRPα) axis dictates the fate of ingested DNA in DCs for immune evasion. Although macrophages were more potent in uptaking tumor DNA, increase of DNA sensing by blocking the interaction of SIRPα with CD47 preferentially occurred in dendritic cells (DCs) but not in macrophages. Mechanistically, CD47 blockade enabled the activation of NADPH oxidase NOX2 in DCs, which in turn inhibited phagosomal acidification and reduced the degradation of tumor mitochondrial DNA (mtDNA) in DCs. mtDNA was recognized by cyclic-GMP-AMP synthase (cGAS) in the DC cytosol, contributing to type I interferon (IFN) production and antitumor adaptive immunity. Thus, our findings have demonstrated how tumor cells inhibit innate sensing in DCs and suggested that the CD47-SIRPα axis is critical for DC-driven antitumor immunity.


Subject(s)
Antigens, Differentiation/metabolism , Colonic Neoplasms/immunology , DNA, Mitochondrial/immunology , Dendritic Cells/immunology , Membrane Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Antibodies, Blocking/therapeutic use , CD47 Antigen/immunology , CD47 Antigen/metabolism , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Cross-Priming , Disease Models, Animal , Humans , Interferon Type I/metabolism , Macrophages/immunology , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Tumor Escape
SELECTION OF CITATIONS
SEARCH DETAIL