Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.839
Filter
1.
J Ethnopharmacol ; 336: 118661, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39159837

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Shuangdan Jiedu Decoction (SJD) is a formula composed of six Chinese herbs with heat-removing and detoxifying, antibacterial, and anti-inflammatory effects, which is clinically used in the therapy of various inflammatory diseases of the lungs including COVID-19, but the therapeutic material basis of its action as well as its molecular mechanism are still unclear. AIM OF THE STUDY: The study attempted to determine the therapeutic effect of SJD on LPS-induced acute lung injury (ALI), as well as to investigate its mechanism of action and assess its therapeutic potential for the cure of inflammation-related diseases in the clinical setting. MATERIALS AND METHODS: We established an ALI model by tracheal drip LPS, and after the administration of SJD, we collected the bronchoalveolar lavage fluid (BALF) and lung tissues of mice and examined the expression of inflammatory factors in them. In addition, we evaluated the effects of SJD on the cyclic guanosine monophosphate-adenosine monophosphate synthase -stimulator of interferon genes (cGAS-STING) and inflammasome by immunoblotting and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS: We demonstrated that SJD was effective in alleviating LPS-induced ALI by suppressing the levels of pro-inflammatory cytokines in the BALF, improving the level of lung histopathology and the number of neutrophils, as well as decreasing the inflammatory factor-associated gene expression. Importantly, we found that SJD could inhibit multiple stimulus-driven activation of cGAS-STING and inflammasome. Further studies showed that the Chinese herbal medicines in SJD had no influence on the cGAS-STING pathway and inflammasome alone at the formulated dose. By increasing the concentration of these herbs, we observed inhibitory effects on the cGAS-STING pathway and inflammasome, and the effect exerted was maximal when the six herbs were combined, indicating that the synergistic effects among these herbs plays a crucial role in the anti-inflammatory effects of SJD. CONCLUSIONS: Our research demonstrated that SJD has a favorable protective effect against ALI, and its mechanism of effect may be associated with the synergistic effect exerted between six Chinese medicines to inhibit the cGAS-STING and inflammasome abnormal activation. These results are favorable for the wide application of SJD in the clinic as well as for the development of drugs for ALI from herbal formulas.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Inflammasomes , Lipopolysaccharides , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Lipopolysaccharides/toxicity , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Nucleotidyltransferases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Male , Signal Transduction/drug effects , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Lung/drug effects , Lung/pathology , Lung/metabolism , Bronchoalveolar Lavage Fluid/cytology
2.
J Ethnopharmacol ; 336: 118741, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39197801

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Amyotrophic lateral sclerosis (ALS) is a fetal neuromuscular disorder characterized by the gradual deterioration of motor neurons. Semen Strychni pulveratum (SSP), a processed version of Semen Strychni (SS) powder, is widely used to treat ALS in China. Vomicine is one of the most primary components of SS. However, their pharmacological effects and mechanisms for ALS remain elusive. AIM OF THE STUDY: This study aimed to evaluate the neuroprotective and anti-neuroinflammatory effects of SSP and vomicine, as well as to explore their protective roles in ALS and the underlying mechanisms. MATERIALS AND METHODS: In vivo, 8-week-old hSOD1-WT mice and hSOD1-G93A mice were orally administered different concentrations of SSP (SSP-L = 5.46 mg/ml, SSP-M = 10.92 mg/ml or SSP-H = 16.38 mg/ml) once every other day for 8 weeks. A series of experiments, including body weight measurement, footprint tests, Hematoxylin & Eosin staining, and Nissl staining, were performed to evaluate the preventive effect of SSP. Immunofluorescence staining, western blotting, and RT-qPCR were subsequently performed to evaluate activation of the cGAS-STING-TBK1 pathway in the spinal cord. In vitro, hSOD1G93A NSC-34 cells were treated with vomicine to further explore the pharmacological mechanism of vomicine in the treatment of ALS via the cGAS-STING-TBK1 pathway. RESULTS: SSP improved motor function, body weight loss, gastrocnemius muscle atrophy, and motor neuron loss in the spine and cortex of hSOD1-G93A mice. Furthermore, the cGAS-STING-TBK1 pathway was activated in the spinal cord of hSOD1-G93A mice, with activation predominantly observed in neurons and microglia. However, the levels of cGAS, STING, and pTBK1 proteins and cGAS, IRF3, IL-6, and IL-1ß mRNA were reversed following intervention with SSP. Vomicine not only downregulated the levels of cGAS, TBK1, IL-6 and IFN-ß mRNA, but also the levels of cGAS and STING protein in hSOD1G93A NSC-34 cells. CONCLUSION: This study demonstrated that SSP and vomicine exert neuroprotective and anti-neuroinflammatory effects in the treatment of ALS. SSP and vomicine may reduce neuroinflammation by regulating the cGAS-STING-TBK1 pathway, and could thereby play a role in ALS treatment.


Subject(s)
Amyotrophic Lateral Sclerosis , Membrane Proteins , Neuroprotective Agents , Nucleotidyltransferases , Protein Serine-Threonine Kinases , Animals , Protein Serine-Threonine Kinases/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Mice , Membrane Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nucleotidyltransferases/metabolism , Male , Signal Transduction/drug effects , Mice, Transgenic , Neuroinflammatory Diseases/drug therapy , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Disease Models, Animal
3.
Biomaterials ; 313: 122766, 2025 Feb.
Article in English | MEDLINE | ID: mdl-39180916

ABSTRACT

The immune resistance of tumor microenvironment (TME) causes immune checkpoint blockade therapy inefficient to hepatocellular carcinoma (HCC). Emerging strategies of using chemotherapy regimens to reverse the immune resistance provide the promise for promoting the efficiency of immune checkpoint inhibitors. The induction of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) in tumor cells evokes the adaptive immunity and remodels the immunosuppressive TME. In this study, we report that mitoxantrone (MIT, a chemotherapeutic drug) activates the cGAS-STING signaling pathway of HCC cells. We provide an approach to augment the efficacy of MIT using a signal transducer and activator of transcription 3 (STAT3) inhibitor called napabucasin (NAP). We prepare an aminoethyl anisamide (AEAA)-targeted polyethylene glycol (PEG)-modified poly (lactic-co-glycolic acid) (PLGA)-based nanocarrier for co-delivery of MIT and NAP. The resultant co-nanoformulation can elicit the cGAS-STING-based immune responses to reshape the immunoresistant TME in the mice orthotopically grafted with HCC. Consequently, the resultant co-nanoformulation can promote anti-PD-1 antibody for suppressing HCC development, generating long-term survival, and inhibiting tumor recurrence. This study reveals the potential of MIT to activate the cGAS-STING signaling pathway, and confirms the feasibility of nano co-delivery for MIT and NAP on achieving HCC chemo-immunotherapy.


Subject(s)
Carcinoma, Hepatocellular , Immunotherapy , Liver Neoplasms , Membrane Proteins , Mitoxantrone , Nucleotidyltransferases , STAT3 Transcription Factor , Mitoxantrone/pharmacology , Mitoxantrone/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Mice , Immunotherapy/methods , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Benzofurans , Naphthoquinones
4.
Methods Mol Biol ; 2854: 117-125, 2025.
Article in English | MEDLINE | ID: mdl-39192124

ABSTRACT

Beyond its role as the bearer of genetic material, DNA also plays a crucial role in the activation phase of innate immunity. Pathogen recognition receptors (PRRs) and their homologs, pathogen-associated molecular patterns (PAMPs), form the foundation for driving innate immune activation and the induction of immune responses during infection. In the context of DNA viruses or bacterial infections, specific DNA sequences are recognized and bound by DNA sensors, marking the DNA as a PAMP for host recognition and subsequent activation of innate immunity. The primary DNA sensor pathway known to date is cGAS-STING, which can induce Type I interferons (IFN) and innate immune responses against viruses and bacteria. Additionally, the cGAS-STING pathway has been identified to mediate functions in autophagy and senescence. Herein, we introduce methods for using DNA PAMPs as molecular tools to study the role of cGAS-STING and its signaling pathway in regulating innate immunity, both in vitro and in vivo.


Subject(s)
DNA , Immunity, Innate , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , DNA/metabolism , DNA/genetics , Animals , Pathogen-Associated Molecular Pattern Molecules/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Mice
5.
Methods Mol Biol ; 2854: 61-74, 2025.
Article in English | MEDLINE | ID: mdl-39192119

ABSTRACT

With the rapid development of CRISPR-Cas9 technology, gene editing has become a powerful tool for studying gene function. Specifically, in the study of the mechanisms by which natural immune responses combat viral infections, gene knockout mouse models have provided an indispensable platform. This article describes a detailed protocol for constructing gene knockout mice using the CRISPR-Cas9 system. This field focuses on the design of single-guide RNAs (sgRNAs) targeting the antiviral immune gene cGAS, embryo microinjection, and screening and verification of gene editing outcomes. Furthermore, this study provides methods for using cGAS gene knockout mice to analyze the role of specific genes in natural immune responses. Through this protocol, researchers can efficiently generate specific gene knockout mouse models, which not only helps in understanding the functions of the immune system but also offers a powerful experimental tool for exploring the mechanisms of antiviral innate immunity.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Immunity, Innate , Mice, Knockout , RNA, Guide, CRISPR-Cas Systems , Animals , Immunity, Innate/genetics , Mice , RNA, Guide, CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Knockout Techniques/methods , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Virus Diseases/immunology , Virus Diseases/genetics
6.
Methods Mol Biol ; 2854: 153-170, 2025.
Article in English | MEDLINE | ID: mdl-39192127

ABSTRACT

cGAS is a key cytosolic dsDNA receptor that senses viral infection and elicits interferon production through the cGAS-cGAMP-STING axis. cGAS is activated by dsDNA from viral and bacterial origins as well as dsDNA leaked from damaged mitochondria and nucleus. Eventually, cGAS activation launches the cell into an antiviral state to restrict the replication of both DNA and RNA viruses. Throughout the long co-evolution, viruses devise many strategies to evade cGAS detection or suppress cGAS activation. We recently reported that the Dengue virus protease NS2B3 proteolytically cleaves human cGAS in its N-terminal region, effectively reducing cGAS binding to DNA and consequent production of the second messenger cGAMP. Several other RNA viruses likely adopt the cleavage strategy. Here, we describe a protocol for the purification of recombinant human cGAS and Dengue NS2B3 protease, as well as the in vitro cleavage assay.


Subject(s)
Dengue Virus , Nucleotidyltransferases , Viral Nonstructural Proteins , Humans , Viral Nonstructural Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Proteolysis , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Nucleotides, Cyclic/metabolism , Dengue/virology , Dengue/metabolism
7.
Nat Commun ; 15(1): 7719, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231966

ABSTRACT

Nucleotidyltransferases (NTases) control diverse physiological processes, including RNA modification, DNA replication and repair, and antibiotic resistance. The Mycobacterium tuberculosis NTase toxin family, MenT, modifies tRNAs to block translation. MenT toxin activity can be stringently regulated by diverse MenA antitoxins. There has been no unifying mechanism linking antitoxicity across MenT homologues. Here we demonstrate through structural, biochemical, biophysical and computational studies that despite lacking kinase motifs, antitoxin MenA1 induces auto-phosphorylation of MenT1 by repositioning the MenT1 phosphoacceptor T39 active site residue towards bound nucleotide. Finally, we expand this predictive model to explain how unrelated antitoxin MenA3 is similarly able to induce auto-phosphorylation of cognate toxin MenT3. Our study reveals a conserved mechanism for the control of tuberculosis toxins, and demonstrates how active site auto-phosphorylation can regulate the activity of widespread NTases.


Subject(s)
Catalytic Domain , Mycobacterium tuberculosis , Nucleotidyltransferases , Phosphorylation , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Models, Molecular , RNA, Transfer/metabolism , RNA, Transfer/genetics , Crystallography, X-Ray
8.
Front Immunol ; 15: 1451305, 2024.
Article in English | MEDLINE | ID: mdl-39295867

ABSTRACT

The cGAS-STING signaling pathway plays a critical role in innate immunity and defense against viral infections by orchestrating intracellular and adaptive immune responses to DNA. In the context of head and neck squamous cell carcinoma (HNSCC), this pathway has garnered significant attention due to its potential relevance in disease development and progression. HNSCC is strongly associated with risk factors such as smoking, heavy alcohol consumption, and human papillomavirus (HPV) infection. The presence or absence of HPV in HNSCC patients has been shown to have a profound impact on patient survival and prognosis, possibly due to the distinct biological characteristics of HPV-associated tumors. This review aims to provide a comprehensive overview of the current therapeutic approaches and challenges in HNSCC management, as well as the involvement of cGAS-STING signaling and its potential in the therapy of HNSCC. In addition, by advancing the present understanding of the mechanisms underlying this pathway, Activation of cGAS-STING-dependent inflammatory signaling downstream of chromosomal instability can exert both anti-tumoral and pro-tumoral effects in a cell-intrinsic manner, suggesting individualized therapy is of great importance. However, further exploration of the cGAS-STING signaling pathway is imperative for the effective management of HNSCC.


Subject(s)
Head and Neck Neoplasms , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Humans , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/etiology , Animals , Papillomavirus Infections/immunology , Papillomavirus Infections/complications
9.
Proc Natl Acad Sci U S A ; 121(39): e2415648121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39297679

ABSTRACT

The 2024 Albert Lasker Basic Medical Research Award was attributed to Zhijian (James) Chen for "the discovery of the cGAS enzyme that senses foreign and self DNA, solving the mystery of how DNA stimulates immune and inflammatory responses." Bringing to bear an ingenious in vitro complementation system, an astute insight, and superlative biochemistry, Chen and colleagues identified cGAS (cGAMP synthase) as both the molecule that perceives cytosolic DNA in infected, stressed, or dying cells and the enzyme that catalyzes the synthesis of cGAMP (cyclic GMP-AMP), a critical second messenger along the route to inflammatory cytokine production. These findings cleared up the reigning confusion surrounding a major mechanism of inciting the innate immune system, with therapeutic implications for fighting infections, containing tumors, and extinguishing autoimmune and inflammatory diseases.


Subject(s)
Cytosol , DNA , Immunity, Innate , Nucleotidyltransferases , Humans , DNA/metabolism , Cytosol/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Animals , Nucleotides, Cyclic/metabolism
10.
Int J Biol Sci ; 20(12): 4713-4730, 2024.
Article in English | MEDLINE | ID: mdl-39309425

ABSTRACT

Extensive loss of alveolar epithelial cells (AECs) undergoing necroptosis is a crucial mechanism of acute lung injury (ALI), but its triggering mechanism needs to be thoroughly investigated. Neutrophil extracellular traps (NETs) play a significant role in ALI. However, the effect of NETs on AECs' death has not been clarified. Our study found that intratracheal instillation of NETs disrupted lung tissue structure, suggesting that NETs could induce ALI in mice. Moreover, we observed that NETs could trigger necroptosis of AECs in vivo and in vitro. The phosphorylation levels of RIPK3 and MLKL were increased in MLE12 cells after NETs treatment (P < 0.05). Mechanistically, NETs taken up by AECs through endocytosis activated the cGAS-STING pathway and triggered AECs necroptosis. The expression of cGAS, STING, TBK1 and IRF3 were increased in MLE12 cells treated with NETs (P < 0.05). Furthermore, the cGAS inhibitor RU.521 inhibited NETs-triggered AECs necroptosis and alleviated the pulmonary damage induced by NETs in mice. In conclusion, our study demonstrates that NETs taken up by AECs via endocytosis can activate the cGAS-STING pathway and trigger AECs necroptosis to promote ALI in mice. Our findings indicate that targeting the NETs/cGAS-STING/necroptosis pathway in AECs is an effective strategy for treating ALI.


Subject(s)
Acute Lung Injury , Alveolar Epithelial Cells , Extracellular Traps , Membrane Proteins , Necroptosis , Nucleotidyltransferases , Animals , Extracellular Traps/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Mice , Nucleotidyltransferases/metabolism , Alveolar Epithelial Cells/metabolism , Membrane Proteins/metabolism , Male , Signal Transduction , Mice, Inbred C57BL , Neutrophils/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
11.
Front Endocrinol (Lausanne) ; 15: 1432819, 2024.
Article in English | MEDLINE | ID: mdl-39301315

ABSTRACT

Maintaining a well-functioning mitochondrial network through the mitochondria quality control (MQC) mechanisms, including biogenesis, dynamics and mitophagy, is crucial for overall health. Mitochondrial dysfunction caused by oxidative stress and further exacerbated by impaired quality control can trigger inflammation through the release of the damage-associated molecular patterns (mtDAMPs). mtDAMPs act by stimulating the cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway. Recently, aberrant signalling of the cGAS-STING axis has been recognised to be closely associated with several sterile inflammatory diseases (e.g. non-alcoholic fatty liver disease, obesity). This may fit the pathophysiology of hypothyroidism, an endocrine disorder characterised by the reduction of thyroid hormone production associated with impaired metabolic fluxes, oxidative balance and inflammatory status. Both 3,5,3'-triiodo-L-tyronine (T3) and its derivative 3,5-diiodo-L-thyronine (3,5-T2), are known to mitigate processes targeting mitochondria, albeit the underlying mechanisms are not yet fully understood. Therefore, we used a chemically induced hypothyroidism rat model to investigate the effect of 3,5-T2 or T3 administration on inflammation-related factors (inflammatory cytokines, hepatic cGAS-STING pathway), oxidative stress, antioxidant defence enzymes, mitochondrial DNA (mtDNA) damage, release and repair, and the MQC system in the liver. Hypothyroid rats showed: i) increased oxidative stress, ii) accumulation of mtDNA damage, iii) high levels of circulating cytokines, iv) hepatic activation of cGAS-STING pathways and v) impairment of MQC mechanisms and autophagy. Both iodothyronines restored oxidative balance by enhancing antioxidant defence, preventing mtDNA damage through the activation of mtDNA repair mechanisms (OGG1, APE1, and POLγ) and promoting autophagy progression. Concerning MQC, both iodothyronines stimulated mitophagy and dynamics, with 3,5-T2 activating fusion and T3 modulating both fusion and fission processes. Moreover, only T3 enhanced mitochondrial biogenesis. Notably, 3,5-T2, but not T3, reversed the hypothyroidism-induced activation of the cGAS-STING inflammatory cascade. In addition, it is noteworthy that 3,5-T2 seems more effective than T3 in reducing circulating pro-inflammatory cytokines IL-6 and IL-1B and in stimulating the release of IL-10, a known anti-inflammatory cytokine. These findings reveal novel molecular mechanisms of hepatic signalling pathways involved in hypothyroidism, which could be targeted by natural iodothyronines, particularly 3,5-T2, paving the way for the development of new treatment strategies for inflammatory diseases.


Subject(s)
Diiodothyronines , Hypothyroidism , Inflammation , Liver , Membrane Proteins , Nucleotidyltransferases , Oxidative Stress , Animals , Rats , Hypothyroidism/metabolism , Hypothyroidism/chemically induced , Hypothyroidism/drug therapy , Hypothyroidism/pathology , Liver/metabolism , Liver/drug effects , Liver/pathology , Nucleotidyltransferases/metabolism , Male , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Diiodothyronines/pharmacology , Membrane Proteins/metabolism , Oxidative Stress/drug effects , Triiodothyronine , Mitochondria/metabolism , Mitochondria/drug effects , Rats, Wistar , Mitochondria, Liver/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/pathology , Signal Transduction/drug effects
12.
Cytokine ; 183: 156737, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39217915

ABSTRACT

BACKGROUND: Opioid activation of the microglia or macrophage Toll-like receptor 4 (TLR4) and associated inflammatory cytokine release are implicated in opioid-induced hyperalgesia and tolerance. The cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS-STING) signaling pathway, activated by double-stranded DNA including mitochondrial DNA (mtDNA), has emerged as another key mediator of inflammatory responses. This study tested the hypothesis that morphine induces immune inflammatory responses in microglia and macrophages involving TLR4 and cGAS-STING pathway. METHODS: BV2 microglia and Raw 264.7 (Raw) macrophage cells were exposed to morphine with and without a STING inhibitor (C176) for 6 h or TLR 4 inhibitor (TAK242) for 24 h. Western blotting and RT-qPCR analyses assessed TLR4, cGAS, STING, nuclear factor-kappa B (NF-κB), and pro-inflammatory cytokine expression. Morphine-induced mitochondria dysfunction was quantified by reactive oxygen species (ROS) release using MitoSOX, mtDNA release by immunofluorescence, and RT-qPCR. Polarization of BV2 and Raw cells was assessed by inducible nitric oxide (iNOS) and CD86 expression. The role of mtDNA on morphine-related inflammation was investigated by mtDNA depletion of the cells with ethidium bromide (EtBr) or cell transfection of mtDNA extracted from morphine-treated cells. RESULTS: Morphine significantly increased the expression of TLR4, cGAS, STING, p65 NF-κB, and cytokines (IL-6 and TNF-α) in BV2 and Raw cells. Morphine-induced mitochondrial dysfunction by increased ROS and mtDNA release; the increased iNOS and CD86 evidenced inflammatory M1-like phenotype polarization. TLR4 and STING inhibitors reduced morphine-induced cytokine release in both cell types. The transfection of mtDNA activated inflammatory signaling proteins, cytokine release, and polarization. Conversely, mtDNA depletion led to the reversal of these effects. CONCLUSION: Morphine activates the cGAS-STING pathway in macrophage cell types. Inhibition of the STING pathway can be an additional method to overcome immune cell inflammation-related morphine tolerance and opioid-induced hyperalgesia.


Subject(s)
Inflammation , Macrophages , Membrane Proteins , Morphine , Nucleotidyltransferases , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Animals , Mice , Signal Transduction/drug effects , Morphine/pharmacology , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Inflammation/metabolism , RAW 264.7 Cells , Macrophages/metabolism , Macrophages/drug effects , DNA, Mitochondrial/metabolism , Microglia/metabolism , Microglia/drug effects , Cytokines/metabolism , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , NF-kappa B/metabolism , Cell Line
13.
J Med Chem ; 67(18): 16235-16247, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39250558

ABSTRACT

Triggering ferroptosis represents a promising anticancer therapeutic strategy, but the development of a selective ferroptosis inducer for cancer-specific therapy remains a great challenge. Herein, a H2S-responsive iridium(III) complex NA-Ir has been well-designed as a ferroptosis inducer. NA-Ir could selectively light up H2S-rich cancer cells, primarily localize in mitochondria, intercalate into mitochondrial DNA (mtDNA), and induce mtDNA damage, exhibiting higher anticancer activity under light irradiation. Mechanistic studies showed that NA-Ir-mediated PDT triggered lipid peroxidation and glutathione peroxidase 4 downregulation through ROS production and GSH depletion, resulting in ferroptosis through multiple pathways. Moreover, the intense mtDNA damage can activate the cyclic GMP-AMP synthase-stimulator of the interferon gene (cGAS-STING) pathway, leading to ferritinophagy and further ferroptosis. RNA-sequencing analysis showed that NA-Ir-mediated PDT mainly affects the expression of genes related to ferroptosis, autophagy, and cancer immunity. This study demonstrates the first cancer-specific example with ferroptosis and cGAS-STING activation, which provides a new strategy for multimodal synergistic therapy.


Subject(s)
Ferroptosis , Iridium , Membrane Proteins , Nucleotidyltransferases , Ferroptosis/drug effects , Humans , Iridium/chemistry , Iridium/pharmacology , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Cell Line, Tumor , Mice , DNA, Mitochondrial/metabolism , Reactive Oxygen Species/metabolism
14.
Proc Natl Acad Sci U S A ; 121(38): e2411747121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39254994

ABSTRACT

Detection of cytosolic DNA by the cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway provides immune defense against pathogens and cancer but can also cause autoimmunity when overactivated. The exonuclease three prime repair exonuclease 1 (TREX1) degrades DNA in the cytosol and prevents cGAS activation by self-DNA. Loss-of-function mutations of the TREX1 gene are linked to autoimmune diseases such as Aicardi-Goutières syndrome, and mice deficient in TREX1 develop lethal inflammation in a cGAS-dependent manner. In order to determine the type of cells in which cGAS activation drives autoinflammation, we generated conditional cGAS knockout mice on the Trex1-/- background. Here, we show that genetic ablation of the cGAS gene in classical dendritic cells (cDCs), but not in macrophages, was sufficient to rescue Trex1-/- mice from all observed disease phenotypes including lethality, T cell activation, tissue inflammation, and production of antinuclear antibodies and interferon-stimulated genes. These results show that cGAS activation in cDC causes autoinflammation in response to self-DNA accumulated in the absence of TREX1.


Subject(s)
Autoimmunity , Dendritic Cells , Exodeoxyribonucleases , Mice, Knockout , Nucleotidyltransferases , Phosphoproteins , Animals , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/deficiency , Dendritic Cells/immunology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/immunology , Mice , Autoimmunity/immunology , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/pathology , Inflammation/immunology , Inflammation/genetics , Macrophages/immunology , Macrophages/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics
15.
Proc Natl Acad Sci U S A ; 121(38): e2400781121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39259589

ABSTRACT

During homeostasis, the endoplasmic reticulum (ER) maintains productive transmembrane and secretory protein folding that is vital for proper cellular function. The ER-resident HSP70 chaperone, binding immunoglobulin protein (BiP), plays a pivotal role in sensing ER stress to activate the unfolded protein response (UPR). BiP function is regulated by the bifunctional enzyme filamentation induced by cyclic-AMP domain protein (FicD) that mediates AMPylation and deAMPylation of BiP in response to changes in ER stress. AMPylated BiP acts as a molecular rheostat to regulate UPR signaling, yet little is known about the molecular consequences of FicD loss. In this study, we investigate the role of FicD in mouse embryonic fibroblast (MEF) response to pharmacologically and metabolically induced ER stress. We find differential BiP AMPylation signatures when comparing robust chemical ER stress inducers to physiological glucose starvation stress and recovery. Wildtype MEFs respond to pharmacological ER stress by down-regulating BiP AMPylation. Conversely, BiP AMPylation in wildtype MEFs increases upon metabolic stress induced by glucose starvation. Deletion of FicD results in widespread gene expression changes under baseline growth conditions. In addition, FicD null MEFs exhibit dampened UPR signaling, altered cell stress recovery response, and unconstrained protein secretion. Taken together, our findings indicate that FicD is important for tampering UPR signaling, stress recovery, and the maintenance of secretory protein homeostasis.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Fibroblasts , Glucose , Unfolded Protein Response , Animals , Mice , Embryo, Mammalian/metabolism , Embryo, Mammalian/cytology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Fibroblasts/metabolism , Glucose/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Mice, Knockout , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Signal Transduction
16.
J Nanobiotechnology ; 22(1): 535, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227944

ABSTRACT

BACKGROUND: Manganese ions (Mn2+) combined with adjuvants capable of damaging and lysing tumor cells form an antitumor nano-modulator that enhances the immune efficacy of cancer therapy through the cascade activation of the cyclic GMP-AMP interferon gene synthase-stimulator (cGAS-STING) pathway, which underscores the importance of developing antitumor nano-modulators, which induce DNA damage and augment cGAS-STING activity, as a critical future research direction. METHODS AND RESULTS: We have successfully synthesized an antitumor nano-modulator, which exhibits good dispersibility and biosafety. This nano-modulator is engineered by loading manganese dioxide nanosheets (M-NS) with zebularine (Zeb), known for its immunogenicity-enhancing effects, and conducting targeted surface modification using hyaluronic acid (HA). After systemic circulation to the tumor site, Mn2+, Zeb, and reactive oxygen species (ROS) are catalytically released in the tumor microenvironment by H+ and H2O2. These components can directly or indirectly damage the DNA or mitochondria of tumor cells, thereby inducing programmed cell death. Furthermore, they promote the accumulation of double-stranded DNA (dsDNA) in the cytoplasm, enhancing the activation of the cGAS-STING signalling pathway and boosting the production of type I interferon and the secretion of pro-inflammatory cytokines. Additionally, Zeb@MH-NS enhances the maturation of dendritic cells, the infiltration of cytotoxic T lymphocytes, and the recruitment of natural killer cells at the tumor site. CONCLUSIONS: This HA-modified manganese-based hybrid nano-regulator can enhance antitumor therapy by boosting innate immune activity and may provide new directions for immunotherapy and clinical translation in cancer.


Subject(s)
Immunity, Innate , Manganese Compounds , Membrane Proteins , Nucleotidyltransferases , Oxides , Signal Transduction , Tumor Microenvironment , Nucleotidyltransferases/metabolism , Tumor Microenvironment/drug effects , Immunity, Innate/drug effects , Animals , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Membrane Proteins/metabolism , Signal Transduction/drug effects , Mice , Oxides/chemistry , Oxides/pharmacology , Manganese/chemistry , Manganese/pharmacology , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Female , Mice, Inbred C57BL
17.
Sci Rep ; 14(1): 20859, 2024 09 06.
Article in English | MEDLINE | ID: mdl-39242811

ABSTRACT

Mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) gastric cancer (GC) exhibits an immune-active tumor microenvironment (TME) compared to MMR proficient (pMMR)/microsatellite stable/Epstein-Barr virus-negative [EBV (-)] GC. The tumor cell-intrinsic cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has been considered a key regulator of immune cell activation in the TME. However, its significance in regulating the immune-active TME in dMMR/MSI-H GC remains unclear. Here, we demonstrated that tumor cell-intrinsic cGAS-STING was highly expressed in dMMR GC compared to pMMR/EBV (-) GC. The expression of tumor cell-intrinsic STING was significantly and positively associated with the number of CD8+ tumor-infiltrating lymphocytes in GC. Analysis of TCGA datasets revealed that the expression of interferon-stimulated genes and STING downstream T-cell attracting chemokines was significantly higher in MSI-H GC compared to other subtypes of GC with EBV (-). These results suggest that tumor cell-intrinsic STING signaling plays a key role in activating immune cells in the dMMR/MSI-H GC TME and might serve as a novel biomarker predicting the efficacy of immunotherapy for GC treatment.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Membrane Proteins , Microsatellite Instability , Signal Transduction , Stomach Neoplasms , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Tumor Microenvironment/immunology , Male , Female , DNA Mismatch Repair/genetics , Middle Aged , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Gene Expression Regulation, Neoplastic , Aged
18.
Nat Commun ; 15(1): 7930, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256398

ABSTRACT

Immune checkpoint blockade (ICB) has emerged as a promising therapeutic option for hepatocellular carcinoma (HCC), but resistance to ICB occurs and patient responses vary. Here, we uncover protein arginine methyltransferase 3 (PRMT3) as a driver for immunotherapy resistance in HCC. We show that PRMT3 expression is induced by ICB-activated T cells via an interferon-gamma (IFNγ)-STAT1 signaling pathway, and higher PRMT3 expression levels correlate with reduced numbers of tumor-infiltrating CD8+ T cells and poorer response to ICB. Genetic depletion or pharmacological inhibition of PRMT3 elicits an influx of T cells into tumors and reduces tumor size in HCC mouse models. Mechanistically, PRMT3 methylates HSP60 at R446 to induce HSP60 oligomerization and maintain mitochondrial homeostasis. Targeting PRMT3-dependent HSP60 methylation disrupts mitochondrial integrity and increases mitochondrial DNA (mtDNA) leakage, which results in cGAS/STING-mediated anti-tumor immunity. Lastly, blocking PRMT3 functions synergize with PD-1 blockade in HCC mouse models. Our study thus identifies PRMT3 as a potential biomarker and therapeutic target to overcome immunotherapy resistance in HCC.


Subject(s)
Carcinoma, Hepatocellular , Chaperonin 60 , Liver Neoplasms , Membrane Proteins , Nucleotidyltransferases , Protein-Arginine N-Methyltransferases , Signal Transduction , Animals , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Humans , Mice , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Chaperonin 60/metabolism , Chaperonin 60/genetics , Cell Line, Tumor , Methylation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Mice, Inbred C57BL , DNA, Mitochondrial/genetics , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Male
19.
J Immunother Cancer ; 12(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39237260

ABSTRACT

OBJECTIVE: Pancreatic cancer is an incurable malignant disease with extremely poor prognosis and a complex tumor microenvironment. We sought to characterize the role of Annexin A1 (ANXA1) in pancreatic cancer, including its ability to promote efferocytosis and antitumor immune responses. METHODS: The tumor expression of ANXA1 and cleaved Caspase-3 (c-Casp3) and numbers of tumor-infiltrating CD68+ macrophages in 151 cases of pancreatic cancer were examined by immunohistochemistry and immunofluorescence. The role of ANXA1 in pancreatic cancer was investigated using myeloid-specific ANXA1-knockout mice. The changes in tumor-infiltrating immune cell populations induced by ANXA1 deficiency in macrophages were assessed by single-cell RNA sequencing and flow cytometry. RESULTS: ANXA1 expression in pancreatic cancer patient samples correlated with the number of CD68+ macrophages. The percentage of ANXA1+ tumor-infiltrating macrophages negatively correlated with c-Casp3 expression and was significantly associated with worse survival. In mice, myeloid-specific ANXA1 deficiency inhibited tumor growth and was accompanied by the accumulation of apoptotic cells in pancreatic tumor tissue caused by inhibition of macrophage efferocytosis, which was dependent on cGAS-STING pathway-induced type I interferon signaling. ANXA1 deficiency significantly remodeled the intratumoral lymphocyte and macrophage compartments in tumor-bearing mice by increasing the number of effector T cells and pro-inflammatory macrophages. Furthermore, combination therapy of ANXA1 knockdown with gemcitabine and anti-programmed cell death protein-1 antibody resulted in synergistic inhibition of pancreatic tumor growth. CONCLUSION: This research uncovers a novel role of macrophage ANXA1 in pancreatic cancer. ANXA1-mediated regulation of efferocytosis by tumor-associated macrophages promotes antitumor immune response via STING signaling, suggesting potential treatment strategies for pancreatic cancer.


Subject(s)
Annexin A1 , Macrophages , Membrane Proteins , Nucleotidyltransferases , Pancreatic Neoplasms , Tumor Microenvironment , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Animals , Humans , Mice , Tumor Microenvironment/immunology , Annexin A1/metabolism , Annexin A1/genetics , Macrophages/metabolism , Macrophages/immunology , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Signal Transduction , Female , Male , Mice, Knockout , Efferocytosis
20.
Article in English | MEDLINE | ID: mdl-39218134

ABSTRACT

Arsenic is a toxic metal-like element widely used in the pesticide, preservative and semiconductor industries. However, accumulation of arsenic through the food chain can cause serious damage to animal and human health. However, the toxic mechanism of arsenic-induced hepatotoxicity in chickens is not clear, and the present study aimed to investigate the potential role of cGAS-STING and NF-κB pathways on inflammatory injury in chicken liver. In this study, 75 white-feathered broilers were divided into a control group, a low-dose arsenic group (4 mg/kg) and a high-dose arsenic group (8 mg/kg) to investigate the toxic effects of arsenic on chicken liver. In this study, we found that pathological changes such as inflammatory cell infiltration and vesicular degeneration occurred in the liver when exposed to ATO. Crucially, exposure to ATO triggered the cGAS-STING pathway and markedly raised the levels of mRNA and protein expression of cGAS, STING, TBK1, and IRF7. The type I interferon response was also triggered. Simultaneously, STING induced the activation of the conventional NF-κB signaling pathway and stimulated the expression of genes associated with inflammation, such as IL-6, TNF-α and IL-1ß. In summary, the induction of inflammatory responses via cGAS-STING and NF-κB signaling pathways under high ATO exposure provides new ideas for further studies on the toxicological mechanisms of arsenic.


Subject(s)
Arsenic Trioxide , Chickens , Immunity, Innate , Liver , NF-kappa B , Nucleotidyltransferases , Signal Transduction , Animals , Arsenic Trioxide/toxicity , NF-kappa B/metabolism , Immunity, Innate/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/immunology , Signal Transduction/drug effects , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Inflammation/chemically induced , Chemical and Drug Induced Liver Injury , Avian Proteins/metabolism , Avian Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL