Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Cell Mol Immunol ; 20(12): 1403-1412, 2023 12.
Article in English | MEDLINE | ID: mdl-37932533

ABSTRACT

Various cellular stress conditions trigger mitochondrial DNA (mtDNA) release from mitochondria into the cytosol. The released mtDNA is sensed by the cGAS-MITA/STING pathway, resulting in the induced expression of type I interferon and other effector genes. These processes contribute to the innate immune response to viral infection and other stress factors. The deregulation of these processes causes autoimmune diseases, inflammatory metabolic disorders and cancer. Therefore, the cGAS-MITA/STING pathway is a potential target for intervention in infectious, inflammatory and autoimmune diseases as well as cancer. In this review, we focus on the mechanisms underlying the mtDNA-triggered activation of the cGAS-MITA/STING pathway, the effects of the pathway under various physiological and pathological conditions, and advances in the development of drugs that target cGAS and MITA/STING.


Subject(s)
Autoimmune Diseases , Neoplasms , Humans , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Signal Transduction , Immunity, Innate , Nucleotidyltransferases/metabolism , Mitochondria/metabolism , Autoimmune Diseases/pathology , Neoplasms/pathology
2.
EMBO J ; 42(20): e113743, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37661833

ABSTRACT

Mitochondria play essential roles in cancer cell adaptation to hypoxia, but the underlying mechanisms remain elusive. Through mitochondrial proteomic profiling, we here find that the prolyl hydroxylase EglN1 (PHD2) accumulates on mitochondria under hypoxia. EglN1 substrate-binding region in the ß2ß3 loop is responsible for its mitochondrial translocation and contributes to breast tumor growth. Furthermore, we identify AMP-activated protein kinase alpha (AMPKα) as an EglN1 substrate on mitochondria. The EglN1-AMPKα interaction is essential for their mutual mitochondrial translocation. After EglN1 prolyl-hydroxylates AMPKα under normoxia, they rapidly dissociate following prolyl-hydroxylation, leading to their immediate release from mitochondria. In contrast, hypoxia results in constant EglN1-AMPKα interaction and their accumulation on mitochondria, leading to the formation of a Ca2+ /calmodulin-dependent protein kinase 2 (CaMKK2)-EglN1-AMPKα complex to activate AMPKα phosphorylation, ensuring metabolic homeostasis and breast tumor growth. Our findings identify EglN1 as an oxygen-sensitive metabolic checkpoint signaling hypoxic stress to mitochondria through its ß2ß3 loop region, suggesting a potential therapeutic target for breast cancer.


Subject(s)
AMP-Activated Protein Kinases , Breast Neoplasms , Female , Humans , AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Hypoxia , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mitochondria/metabolism , Proteomics
3.
Elife ; 122023 08 17.
Article in English | MEDLINE | ID: mdl-37589705

ABSTRACT

N6-methyladenosine (m6A) methylation of RNA by the methyltransferase complex (MTC), with core components including METTL3-METTL14 heterodimers and Wilms' tumor 1-associated protein (WTAP), contributes to breast tumorigenesis, but the underlying regulatory mechanisms remain elusive. Here, we identify a novel cleaved form METTL3a (residues 239-580 of METTL3). We find that METTL3a is required for the METTL3-WTAP interaction, RNA m6A deposition, as well as cancer cell proliferation. Mechanistically, we find that METTL3a is essential for the METTL3-METTL3 interaction, which is a prerequisite step for recruitment of WTAP in MTC. Analysis of m6A sequencing data shows that depletion of METTL3a globally disrupts m6A deposition, and METTL3a mediates mammalian target of rapamycin (mTOR) activation via m6A-mediated suppression of TMEM127 expression. Moreover, we find that METTL3 cleavage is mediated by proteasome in an mTOR-dependent manner, revealing positive regulatory feedback between METTL3a and mTOR signaling. Our findings reveal METTL3a as an important component of MTC, and suggest the METTL3a-mTOR axis as a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Cell Cycle Proteins , Methyltransferases , RNA Splicing Factors , Humans , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic , Cytoplasm , Methyltransferases/genetics , RNA , RNA Splicing Factors/genetics , Breast Neoplasms/pathology , Disease Progression
4.
J Mol Cell Biol ; 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37442610

ABSTRACT

Estrogen receptor α (ERα) is an important driver and therapeutic target in approximately 70% of breast cancers. How ERα drives breast carcinogenesis is not fully understood. In this study, we show that ERα is a negative regulator of type I interferon (IFN) response, which is critical for breast carcinogenesis. Activation of ERα by its natural ligand estradiol inhibits IFN-ß-induced transcription of downstream IFN-stimulated genes (ISGs), whereas deficiency of ERα or stimulation with its antagonist fulvestrant has opposite effects. Mechanistically, ERα inhibits type I IFN response by two distinct mechanisms. ERα induces expression of the histone 2A variant H2A.Z, which restricts engagement of the IFN-stimulated gene factor 3 (ISGF3) complex at the ISG promoters. ERα also interacts with STAT2, which leads to disruption of the ISGF3 complex. These two events mutually lead to transcriptional inhibition of ISGs induced by type I IFNs. In a xenograft mouse tumor model, fulvestrant enhances the ability of IFN-ß to suppress ERα+ breast tumor growth. Consistently, clinical data suggests that ERα+ breast cancer patients with higher levels of ISGs exhibit an increased survival rate. Our findings suggest that ERα inhibits type I IFN response via two distinct mechanisms to promote breast cancer.

5.
Cell Mol Immunol ; 20(2): 175-188, 2023 02.
Article in English | MEDLINE | ID: mdl-36600052

ABSTRACT

Upon viral infection, cytoplasmic pattern recognition receptors detect viral nucleic acids and activate the adaptor protein VISA/MAVS- or MITA/STING-mediated innate antiviral response. Whether and how the innate antiviral response is regulated by neuronal endocrine functions is unclear. Here, we show that viral infection reduced the serum levels of the ß-adrenergic hormones epinephrine and norepinephrine as well as the cellular levels of their receptors ADRB1 and ADRB2. We further show that an increase in epinephrine/norepinephrine level inhibited the innate antiviral response in an ADRB1-/2-dependent manner. Mechanistically, epinephrine/norepinephrine stimulation activated the downstream kinase PKA, which catalyzed the phosphorylation of MITA at S241, S243 and T263, inhibiting MITA activation and suppressing the innate immune response to DNA virus. In addition, phosphorylation of VISA at T54 by PKA antagonized the innate immune response to RNA virus. These findings reveal the regulatory mechanisms of innate antiviral responses by epinephrine/norepinephrine and provide a possible explanation for increased host susceptibility to viral infection in stressful and anxiety-promoting situations.


Subject(s)
Membrane Proteins , Virus Diseases , Humans , Antiviral Agents , Epinephrine/pharmacology , Immunity, Innate/genetics , Membrane Proteins/genetics , Norepinephrine/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Enzyme Activation , Cyclic AMP-Dependent Protein Kinases/metabolism
6.
Proc Natl Acad Sci U S A ; 119(43): e2207280119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36252040

ABSTRACT

The current view of nucleic acid-mediated innate immunity is that binding of intracellular sensors to nucleic acids is sufficient for their activation. Here, we report that endocytosis of virus or foreign DNA initiates a priming signal for the DNA sensor cyclic GMP-AMP synthase (cGAS)-mediated innate immune response. Mechanistically, viral infection or foreign DNA transfection triggers recruitment of the spleen tyrosine kinase (SYK) and cGAS to the endosomal vacuolar H+ pump (V-ATPase), where SYK is activated and then phosphorylates human cGASY214/215 (mouse cGasY200/201) to prime its activation. Upon binding to DNA, the primed cGAS initiates robust cGAMP production and mediator of IRF3 activation/stimulator of interferon genes-dependent innate immune response. Consistently, blocking the V-ATPase-SYK axis impairs DNA virus- and transfected DNA-induced cGAMP production and expression of antiviral genes. Our findings reveal that V-ATPase-SYK-mediated tyrosine phosphorylation of cGAS following endocytosis of virus or other cargos serves as a priming signal for cGAS activation and innate immune response.


Subject(s)
Endocytosis , Immunity, Innate , Nucleotidyltransferases , Syk Kinase , Vacuolar Proton-Translocating ATPases , Animals , Humans , Mice , DNA , Interferons/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/genetics , Syk Kinase/metabolism , Tyrosine , Vacuolar Proton-Translocating ATPases/metabolism
7.
Signal Transduct Target Ther ; 7(1): 137, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35468896

ABSTRACT

Whether and how innate antiviral response is regulated by humoral metabolism remains enigmatic. We show that viral infection induces progesterone via the hypothalamic-pituitary-adrenal axis in mice. Progesterone induces downstream antiviral genes and promotes innate antiviral response in cells and mice, whereas knockout of the progesterone receptor PGR has opposite effects. Mechanistically, stimulation of PGR by progesterone activates the tyrosine kinase SRC, which phosphorylates the transcriptional factor IRF3 at Y107, leading to its activation and induction of antiviral genes. SARS-CoV-2-infected patients have increased progesterone levels, and which are co-related with decreased severity of COVID-19. Our findings reveal how progesterone modulates host innate antiviral response, and point to progesterone as a potential immunomodulatory reagent for infectious and inflammatory diseases.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antiviral Agents , COVID-19/genetics , Humans , Hypothalamo-Hypophyseal System , Immunity, Innate/genetics , Mice , Pituitary-Adrenal System , Progesterone/pharmacology
8.
Nat Commun ; 13(1): 1700, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35361799

ABSTRACT

Anti-PD-1/PD-L1 immunotherapy has achieved impressive therapeutic outcomes in patients with multiple cancer types. However, the underlined molecular mechanism(s) for moderate response rate (15-25%) or resistance to PD-1/PD-L1 blockade remains not completely understood. Here, we report that inhibiting the deubiquitinase, USP8, significantly enhances the efficacy of anti-PD-1/PD-L1 immunotherapy through reshaping an inflamed tumor microenvironment (TME). Mechanistically, USP8 inhibition increases PD-L1 protein abundance through elevating the TRAF6-mediated K63-linked ubiquitination of PD-L1 to antagonize K48-linked ubiquitination and degradation of PD-L1. In addition, USP8 inhibition also triggers innate immune response and MHC-I expression largely through activating the NF-κB signaling. Based on these mechanisms, USP8 inhibitor combination with PD-1/PD-L1 blockade significantly activates the infiltrated CD8+ T cells to suppress tumor growth and improves the survival benefit in several murine tumor models. Thus, our study reveals a potential combined therapeutic strategy to utilize a USP8 inhibitor and PD-1/PD-L1 blockade for enhancing anti-tumor efficacy.


Subject(s)
Endopeptidases , Endosomal Sorting Complexes Required for Transport , Immunotherapy , Neoplasms , Tumor Microenvironment , Ubiquitin Thiolesterase , Animals , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics
9.
Int J Endocrinol ; 2022: 6283706, 2022.
Article in English | MEDLINE | ID: mdl-35265125

ABSTRACT

Objectives: To analyze and summarize the effect of SSA treatment on EAS due to p-NETs (EAS-p-NETs). Methods: Thirteen patients with EAS-p-NETs treated with SSAs at our center or described in the literature were included in this study. Clinical characteristics, laboratory data, imaging studies, histopathologic results, the effect of SSA treatment, and the prognosis of these EAS-p-NET patients were evaluated. Results: Four males and 9 females with an average age of 42.9 years were included in the study. The mean duration of follow-up was 38.8 ± 28.2 months. As one of the combined treatment measures, SSAs controlled the levels of ACTH and cortisol in 9 of the 13 patients (69.2%). Partial response was observed in 3 patients (23.1%), stable disease in 2 patients (15.4%), and progressive disease in 6 patients (46.2%). The median time to tumor progression was 24 months, and the median overall survival was 61 months. The side effects of SSA treatment included temporary mild abdominal pain, diarrhea, gallstones, and cholecystitis. Conclusions: As a supplemental therapy, SSA treatment led to clinical and biochemical improvement with a good safety profile in patients exhibiting EAS-p-NET with metastasis. However, tumor progression was inhibited by SSA treatment in only a few patients. Combined with other treatments, SSAs may improve the prognosis of patients with EAS-p-NETs.

12.
Cell Mol Immunol ; 18(5): 1186-1196, 2021 05.
Article in English | MEDLINE | ID: mdl-33785841

ABSTRACT

Mitochondrial stress (mitostress) triggered by viral infection or mitochondrial dysfunction causes the release of mitochondrial DNA (mtDNA) into the cytosol and activates the cGAS-mediated innate immune response. The regulation of mtDNA release upon mitostress remains uncharacterized. Here, we identified mitochondria-associated vaccinia virus-related kinase 2 (VRK2) as a key regulator of this process. VRK2 deficiency inhibited the induction of antiviral genes and caused earlier and higher mortality in mice after viral infection. Upon viral infection, VRK2 associated with voltage-dependent anion channel 1 (VDAC1) and promoted VDAC1 oligomerization and mtDNA release, leading to the cGAS-mediated innate immune response. VRK2 was also required for mtDNA release and cGAS-mediated innate immunity triggered by nonviral factors that cause Ca2+ overload but was not required for the cytosolic nucleic acid-triggered innate immune response. Thus, VRK2 plays a crucial role in the mtDNA-triggered innate immune response and may be a potential therapeutic target for infectious and autoimmune diseases associated with mtDNA release.


Subject(s)
Antiviral Agents/metabolism , DNA, Mitochondrial/metabolism , Immunity, Innate , Mitochondria/metabolism , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/deficiency , Voltage-Dependent Anion Channel 1/metabolism
13.
Zhonghua Nan Ke Xue ; 26(10): 917-921, 2020 Nov.
Article in Chinese | MEDLINE | ID: mdl-33382224

ABSTRACT

OBJECTIVE: To evaluate the validity of psychological care combined with enhanced recovery after surgery (PC+ERAS) management in perioperative nursing care of andrological patients. METHODS: A total of 300 male patients undergoing andrological surgery were included in this study, 150 given PC+ERAS and the other 150 receiving routine nursing care as controls. We evaluated anxiety and depression of all the patients on admission and discharge using Self-Rating Anxiety Scale (SAS) and Self-Rating Depression Scale (SDS), and compared post-operative hospital days, off-bed time, first passage of flatus, Visual Analog Scale (VAS) score and satisfaction with nursing care between the two groups of patients. RESULTS: On discharge, significant improvement was observed in SAS and SDS scores in the PC+ERAS group compared with the baseline, even more significant than in the control group (P < 0.01), but no obvious improvement was seen in the controls (P > 0.05). The patients in the PC+ERAS group also achieved a significantly shorter post-operative hospital stay, earlier post-operative off-bed time and passage of flatus, lower VAS score, and higher satisfaction with nursing care than those in the control group (P < 0.05). CONCLUSIONS: Psychological care combined with ERAS management deserves wide application in the perioperative nursing care of andrological patients, which can significantly improve the patients' anxiety and depression, shorten post-operative hospital stay, reduce VAS score, and increase their satisfaction with nursing care.


Subject(s)
Enhanced Recovery After Surgery , Perioperative Nursing , Urologic Surgical Procedures, Male/nursing , Urologic Surgical Procedures, Male/psychology , Humans , Length of Stay , Male , Postoperative Complications , Postoperative Period
14.
Biomark Res ; 8: 36, 2020.
Article in English | MEDLINE | ID: mdl-32874588

ABSTRACT

BACKGROUND: Mixed phenotype acute leukemia (MPAL) is a rare leukemia and is regarded as a high-risk entity with a poor prognosis. Induction therapy of an acute lymphoblastic leukemia type or hybrid regimen and hematopoietic stem cell transplantation has been recommended for MPAL. However, the optimal therapies for relapsed or refractory MPAL remain unclear, especially for relapse after stem cell transplantation. Donor-derived chimeric antigen receptor T (CAR-T) cell therapy may be a promising therapeutic option for patients with MPAL who express target antigens and have relapsed after stem cell transplantation. However, recurrence remains a challenge, and reinfusion of CAR-T cells is not always effective. An infusion of secondary donor-derived humanized CD19-modified CAR-T cells may be effective in inducing remission. CASE PRESENTATION: We report a case of MPAL with CD19 expression. The patient was treated with acute lymphoblastic leukemia-like induction and consolidation therapies but remained positive for SET-NUP214 fusion gene transcript. He subsequently underwent a haploidentical stem cell transplantation but relapsed within 6 months. He then underwent donor-derived CD19-targeted CAR-T cell therapy and achieved a sustained, complete molecular remission. Unfortunately, he developed a CD19-positive relapse after 2 years. Donor-derived humanized CD19-directed CAR-T cells induced a second complete molecular remission without severe cytokine release syndrome or acute graft-versus-host disease. CONCLUSION: This case demonstrated the efficacy and safety of humanized donor-derived CD19-modified CAR-T cell infusion for treating the recurrence of MPAL previously exposed to murine-derived CD19-directed CAR-T cells.

15.
Proc Natl Acad Sci U S A ; 117(35): 21568-21575, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817552

ABSTRACT

The DNA sensor cGMP-AMP synthase (cGAS) senses cytosolic microbial or self DNA to initiate a MITA/STING-dependent innate immune response. cGAS is regulated by various posttranslational modifications at its C-terminal catalytic domain. Whether and how its N-terminal unstructured domain is regulated by posttranslational modifications remain unknown. We identified the acetyltransferase KAT5 as a positive regulator of cGAS-mediated innate immune signaling. Overexpression of KAT5 potentiated viral-DNA-triggered transcription of downstream antiviral genes, whereas a KAT5 deficiency had the opposite effects. Mice with inactivated Kat5 exhibited lower levels of serum cytokines in response to DNA virus infection, higher viral titers in the brains, and more susceptibility to DNA-virus-induced death. Mechanistically, KAT5 catalyzed acetylation of cGAS at multiple lysine residues in its N-terminal domain, which promoted its DNA-binding ability. Our findings suggest that KAT5-mediated cGAS acetylation at its N terminus is important for efficient innate immune response to DNA virus.


Subject(s)
DNA Virus Infections/immunology , DNA Viruses/immunology , Lysine Acetyltransferase 5/immunology , Nucleotidyltransferases/immunology , Acetylation , Animals , Cyclic GMP/metabolism , DNA Virus Infections/genetics , DNA Virus Infections/metabolism , DNA Viruses/genetics , Female , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon-beta/immunology , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Processing, Post-Translational , Signal Transduction , Viral Proteins/metabolism
16.
Cell Discov ; 6: 26, 2020.
Article in English | MEDLINE | ID: mdl-32351706

ABSTRACT

The cyclic GMP-AMP synthase (cGAS) is a widely used DNA sensor, which detects cytosolic DNA species without a preference of self or non-self microbial DNA in interphase to initiate innate immune response. How cGAS is regulated to avoid self-DNA sensing upon nuclear envelope breakdown (NEBD) during mitosis remains enigmatic. Here we show that cGAS is mostly localized in the cytoplasm in interphase and rapidly translocated to chromosomes upon NEBD in mitosis. The major mitotic kinase CDK1-cyclin B complex phosphorylates human cGAS at S305 or mouse cGAS at S291, which inhibits its ability to synthesize cGAMP upon mitotic entry. The type 1 phosphatase PP1 dephosphorylates cGAS upon mitotic exit to enable its DNA sensing ability. Our findings reveal a mechanism on how the DNA sensor cGAS is post-translationally regulated by cell cycle-dependent enzymes to ensure its proper activation for host defense of cytosolic DNA in interphase and inert to self-DNA in mitosis.

17.
Biochem Biophys Res Commun ; 526(1): 199-205, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32201077

ABSTRACT

Upon detection of viral DNA, the cytoplasmic DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS) utilizes GTP and ATP as substrates to synthesize the second messenger molecule 2'3'cyclic GMP-AMP (cGAMP), which binds to the ER-associated adaptor protein MITA/STING to signal innate antiviral response to DNA virus. How the cGAS-MITA pathways are post-translationally regulated is not fully understood. In this study, we identified the tyrosine kinase CSK as a positive regulator of cGAS-MITA mediated innate antiviral response. CSK-deficiency inhibits DNA virus-triggered induction of downstream antiviral effector genes. Following DNA virus infection, CSK phosphorylates MITA at Y240 and Y245, which is important for its activation. These results suggest that CSK plays a role in modulating innate immune response to DNA virus.


Subject(s)
CSK Tyrosine-Protein Kinase/metabolism , DNA Viruses/immunology , Immunity, Innate , Membrane Proteins/metabolism , Animals , CSK Tyrosine-Protein Kinase/deficiency , Cell Line , Humans , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation
18.
Cell Mol Immunol ; 17(11): 1126-1135, 2020 11.
Article in English | MEDLINE | ID: mdl-31511639

ABSTRACT

The mitochondrial virus-induced signaling adaptor (VISA, also called mitochondrial antiviral signaling, MAVS) protein is a central adaptor in the innate immune response to cytosolic viral RNA. Viral infection causes the aggregation of VISA, which is important for its recruitment of downstream signaling components. How VISA aggregation is regulated remains unknown. Here, we found that sorting nexin 8 (SNX8) is a positive regulator of the RNA virus-triggered induction of downstream effector genes and innate immune response. The brains and lungs of Snx8-/- mice infected with RNA viruses exhibited lower serum cytokine levels and higher viral titers than those of wild-type mice, resulting in higher lethality. Mechanistically, viral infection induced the translocation of SNX8 from the cytosol to mitochondria and its increased association with VISA, leading to VISA aggregation, its recruitment of downstream signaling components and the induction of downstream antiviral genes. Our findings suggest that SNX8 is a critical component of the RIG-I-like receptor (RLR)-mediated innate immune response by modulating VISA aggregation and activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate , Protein Aggregates , RNA Viruses/immunology , Sorting Nexins/metabolism , Animals , Cells, Cultured , Female , Male , Mice , Protein Binding , Signal Transduction , Sorting Nexins/genetics
19.
Annu Rev Immunol ; 38: 79-98, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31800327

ABSTRACT

DNA has been known to be a potent immune stimulus for more than half a century. However, the underlying molecular mechanisms of DNA-triggered immune response have remained elusive until recent years. Cyclic GMP-AMP synthase (cGAS) is a major cytoplasmic DNA sensor in various types of cells that detect either invaded foreign DNA or aberrantly located self-DNA. Upon sensing of DNA, cGAS catalyzes the formation of cyclic GMP-AMP (cGAMP), which in turn activates the ER-localized adaptor protein MITA (also named STING) to elicit the innate immune response. The cGAS-MITA axis not only plays a central role in host defense against pathogen-derived DNA but also acts as a cellular stress response pathway by sensing aberrantly located self-DNA, which is linked to the pathogenesis of various human diseases. In this review, we summarize the spatial and temporal mechanisms of host defense to cytoplasmic DNA mediated by the cGAS-MITA axis and discuss the association of malfunctions of this axis with autoimmune and other diseases.


Subject(s)
DNA/immunology , Immunity, Innate , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmunity , Biomarkers , Cytoplasm/immunology , Cytoplasm/metabolism , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Immune Evasion , Interferon Type I/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism
20.
Cell Res ; 29(3): 193-205, 2019 03.
Article in English | MEDLINE | ID: mdl-30651583

ABSTRACT

The mechanisms on metabolic regulation of immune responses are still elusive. We show here that viral infection induces immediate-early NF-κB activation independent of viral nucleic acid-triggered signaling, which triggers a rapid transcriptional induction of bile acid (BA) transporter and rate-limiting biosynthesis enzymes as well as accumulation of intracellular BAs in divergent cell types. The accumulated intracellular BAs activate SRC kinase via the TGR5-GRK-ß-arrestin axis, which mediates tyrosine phosphorylation of multiple antiviral signaling components including RIG-I, VISA/MAVS, MITA/STING, TBK1 and IRF3. The tyrosine phosphorylation of these components by SRC conditions for efficient innate antiviral immune response. Consistently, TGR5 deficiency impairs innate antiviral immunity, whereas BAs exhibit potent antiviral activity in wild-type but not TGR5-deficient cells and mice. Our findings reveal an intrinsic and universal role of intracellular BA metabolism in innate antiviral immunity.


Subject(s)
Bile Acids and Salts/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , HEK293 Cells , Herpesvirus 1, Human/metabolism , Humans , Immunity, Innate/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation/physiology , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...